1
|
Glinka DM, MacGregor GG. The PAR2 Antagonist Larazotide Can Mitigate Acute Histamine-Stimulated Epithelial Barrier Disruption in Keratinocytes: A Potential Adjunct Treatment for Atopic Dermatitis. JID INNOVATIONS 2025; 5:100369. [PMID: 40330848 PMCID: PMC12051560 DOI: 10.1016/j.xjidi.2025.100369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 03/24/2025] [Accepted: 03/24/2025] [Indexed: 05/08/2025] Open
Abstract
Atopic dermatitis (AD) is a chronic inflammatory skin condition with evidence of defects in the barrier properties of the epidermis. Changes in the permeability properties of the tight junction have been reported in AD, and reversing this leaky tight junction may be a potential treatment for AD. This study aimed to determine the effect of larazotide, an antagonist of the protease-activated receptor 2, on the permeability and barrier properties of the tight junctions in keratinocyte monolayers. Normal human epithelial keratinocytes were grown in culture on permeable supports. The effects of larazotide on transepithelial resistance and permeability properties of keratinocyte monolayers were studied before and after histamine challenge. Larazotide mitigated the disruptive effect of histamine on epithelial permeability by increasing the electrical resistance and decreasing epithelial permeability. Larazotide may be beneficial as a topical therapeutic for AD; however, the permeability properties of the short-peptide larazotide through the uppers layers of the epidermis is currently unknown. In conclusion, the protease-activated receptor 2 antagonist larazotide has a protective effect on keratinocyte monolayers and may be useful as an adjunct therapeutic agent to enhance barrier function and promote epidermal healing in AD.
Collapse
Affiliation(s)
| | - Gordon G. MacGregor
- Alabama College of Osteopathic Medicine, Dothan, Alabama, USA
- Orlando College of Osteopathic Medicine, Winter Garden, Florida, USA
- Yogalytes LLC, Huntsville, Alabama, USA
| |
Collapse
|
2
|
Wu Y, Wang XQ, Wu JY, Chen YJ, Bai JX, Li ASM, Fan XY, Wong LY, Wang L, Fu XQ, Yu ZL. A tri-compound formula comprising Ginsenoside Rg1, tetrandrine and icariin alleviates atopic dermatitis symptoms in a mouse model. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 141:156737. [PMID: 40222169 DOI: 10.1016/j.phymed.2025.156737] [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: 05/19/2024] [Revised: 03/16/2025] [Accepted: 04/06/2025] [Indexed: 04/15/2025]
Abstract
BACKGROUND Atopic dermatitis (AD) is characterized by both IgE- and non-IgE-mediated immune responses, as well as skin barrier dysfunction. Ginsenoside Rg1, tetrandrine, and icariin each exhibit distinct properties that may contribute to the management of AD. Ginsenoside Rg1 has demonstrated efficacy in mitigating IgE-mediated allergic rhinitis, while tetrandrine is known to suppress abnormal T-cell activation. Icariin has been shown to improve intestinal barrier function, which is crucial in conditions like AD. However, the potential effectiveness of the combined formula of these compounds, referred to as GTI, in treating AD remains unexplored. PURPOSE This study aimed to investigate the anti-AD effects and mechanisms of GTI in a mouse model. METHODS A calcipotriol (MC903)-induced AD-like dermatitis mouse model was used to evaluate the anti-AD effects of GTI. Dermatitis scores and mouse ear thickness were recorded to assess disease severity. Ear tissues, ear-draining lymph nodes, spleens and sera were collected for use in the investigation of the effects and mechanisms of action of GTI. RESULTS Topical application of GTI significantly alleviated AD-like dermatitis in mice, as evidenced by decreased dermatitis scores, reduced ear thickening, and diminished epidermal and dermal thickness, along with lower levels of the inflammatory cytokines IL-1β and IL-4 in ear tissues. Unlike the positive dexamethasone, GTI had no significant toxicity in the model mice. Topical GTI lowered serum IgE levels and diminished the accumulation of eosinophils and mast cells in ear tissues of model mice, suggesting that GTI mitigates IgE-mediated allergic reactions. GTI significantly decreased the numbers of CD4+ T cells in ear tissues, ear-draining lymph nodes and the spleen, demonstrating its suppressive effect on hyperactive immune responses. The protein levels of ZO-1 and claudin-1, two tight junction proteins, were elevated in the ear tissues of mice treated with GTI, indicating a beneficial effect of this formula on skin barrier function. Additionally, GTI inhibited the activation of mitogen-activated protein kinases (MAPKs), as indicated by the downregulation of phospho-p38 (Thr180/182), phospho-ERK (Thr202/Tyr204), and phospho-JNK (Thr183/185) protein levels in mouse ear tissues. CONCLUSION This study, for the first time, demonstrated that the topical application of GTI alleviates symptoms of AD without overt toxicity in a calcipotriol-induced AD mouse model. The anti-AD effects of GTI are associated with the suppression of allergic reactions, reduction of hyperactive immune responses, improvement of skin barrier function, and inhibition of MAPK activation. These findings suggest that GTI has the potential to be developed into a safe and effective treatment for AD.
Collapse
Affiliation(s)
- Ying Wu
- Centre for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Xiao-Qi Wang
- Centre for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Jia-Ying Wu
- Centre for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Ying-Jie Chen
- Centre for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Jing-Xuan Bai
- Centre for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Amy Sze-Man Li
- Centre for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Xiao-Yun Fan
- Centre for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Lut-Yi Wong
- Centre for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Li Wang
- Centre for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Xiu-Qiong Fu
- Centre for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China.
| | - Zhi-Ling Yu
- Centre for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China; JaneClare Transdermal TCM Therapy Laboratory, Hong Kong Baptist University, Hong Kong SAR, China; Research and Development Centre for Natural Health Products, HKBU Institute for Research and Continuing Education, Shenzhen, China.
| |
Collapse
|
3
|
Meesters LD, Roubroeks JAY, Gerritsen A, Velthuijs N, Klijnhout JA, Laberthonnière C, van Vlijmen-Willems IM, Hübenthal M, Rodijk-Olthuis D, Peters RHW, Rikken G, Szymczak S, Fyhrquist N, Alenius H, Weidinger S, Smits JPH, Mhlanga M, Zhou H, Niehues H, van den Bogaard EH. Dissecting key contributions of T H2 and T H17 cytokines to atopic dermatitis pathophysiology. J Allergy Clin Immunol 2025:S0091-6749(25)00570-6. [PMID: 40409379 DOI: 10.1016/j.jaci.2025.05.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 04/30/2025] [Accepted: 05/08/2025] [Indexed: 05/25/2025]
Abstract
BACKGROUND In atopic dermatitis (AD), epidermal disease hallmarks are driven by a complex cutaneous inflammatory milieu that varies between patients. How these variable inflammatory signals affect cellular and molecular epidermal AD phenotypes is difficult to study in vivo. OBJECTIVE We aimed to unravel which AD-associated cytokines drive specific epidermal disease hallmarks. METHODS We utilized primary and immortalized keratinocyte-derived human epidermal equivalents stimulated with TH2, TH17, and TH22 cytokines. RESULTS Morphologic, functional, and transcriptomic analyses revealed that TH2 cytokines IL-4 and IL-13 were the main inducers of a proinflammatory and hyperproliferative epidermis. The presence of IL-17A or IL-22 in the TH2 milieu, and especially TH2 + IL-22, most closely resembled AD hallmarks including spongiosis, more severe keratinocyte differentiation defects, and epidermal barrier dysfunction. Single-cell spatial transcriptomics showed expansion of keratinocytes expressing high levels of proliferation genes and downregulation of differentiation genes in the upper epidermal layers. The transcriptomic comparison to in vivo AD lesional skin indicated that the TH2 + IL-22 AD model demonstrated greatest resemblance and identified AD disease marker genes altered by TH2 + IL-22 such as downregulated ACER1 and AKR1C3. Gene expression levels were restored by combinatory exposure to the aryl hydrocarbon receptor ligand tapinarof and the Janus kinase inhibitor tofacitinib. This combined therapeutic approach also completely restored epidermal barrier function and improved morphologic disease hallmarks. CONCLUSION Our results reveal the important role of IL-22 in the TH2-driven acute AD pathophysiology and highlight the potential of combinatory medicine in targeted treatment of AD.
Collapse
Affiliation(s)
- Luca D Meesters
- Department of Dermatology, Radboud University Medical Center (Radboudumc), Nijmegen, The Netherlands; Department of Molecular Developmental Biology, Faculty of Science, Radboud University, Nijmegen, The Netherlands
| | - Janou A Y Roubroeks
- Department of Molecular Developmental Biology, Faculty of Science, Radboud University, Nijmegen, The Netherlands
| | - Aranka Gerritsen
- Department of Dermatology, Radboud University Medical Center (Radboudumc), Nijmegen, The Netherlands
| | - Niels Velthuijs
- Department of Cell Biology, Faculty of Science, Radboud University, Nijmegen, The Netherlands
| | - Jaimy A Klijnhout
- Department of Dermatology, Radboud University Medical Center (Radboudumc), Nijmegen, The Netherlands
| | - Camille Laberthonnière
- Department of Molecular Developmental Biology, Faculty of Science, Radboud University, Nijmegen, The Netherlands; Department of Cell Biology, Faculty of Science, Radboud University, Nijmegen, The Netherlands
| | | | - Matthias Hübenthal
- Department of Dermatology, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Diana Rodijk-Olthuis
- Department of Dermatology, Radboud University Medical Center (Radboudumc), Nijmegen, The Netherlands
| | - Rens H W Peters
- Department of Dermatology, Radboud University Medical Center (Radboudumc), Nijmegen, The Netherlands
| | - Gijs Rikken
- Department of Dermatology, Radboud University Medical Center (Radboudumc), Nijmegen, The Netherlands
| | - Silke Szymczak
- Institute of Medical Biometry and Statistics, University of Lübeck, Lübeck, Germany
| | - Nanna Fyhrquist
- Department of Health Sciences, Karlstad University, Karlstad, Sweden; Institute of Environmental Medicine, Karolinska Institute, Stockholm, Sweden
| | - Harri Alenius
- Institute of Environmental Medicine, Karolinska Institute, Stockholm, Sweden; Human Microbiome Research Program, University of Helsinki, Helsinki, Finland
| | - Stephan Weidinger
- Department of Dermatology, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Jos P H Smits
- Department of Dermatology, Radboud University Medical Center (Radboudumc), Nijmegen, The Netherlands; Department of Dermatology, University Hospital Düsseldorf, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
| | - Musa Mhlanga
- Department of Cell Biology, Faculty of Science, Radboud University, Nijmegen, The Netherlands; Department of Human Genetics, Radboudumc, Nijmegen, The Netherlands
| | - Huiqing Zhou
- Department of Molecular Developmental Biology, Faculty of Science, Radboud University, Nijmegen, The Netherlands; Department of Human Genetics, Radboudumc, Nijmegen, The Netherlands
| | - Hanna Niehues
- Department of Dermatology, Radboud University Medical Center (Radboudumc), Nijmegen, The Netherlands
| | - Ellen H van den Bogaard
- Department of Dermatology, Radboud University Medical Center (Radboudumc), Nijmegen, The Netherlands.
| |
Collapse
|
4
|
Chen M, Wang Y, Wang M, Xu S, Tan Z, Cai Y, Xiao X, Wang B, Deng Z, Li J. Keratin 6A promotes skin inflammation through JAK1-STAT3 activation in keratinocytes. J Biomed Sci 2025; 32:47. [PMID: 40346694 PMCID: PMC12065298 DOI: 10.1186/s12929-025-01143-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Accepted: 04/29/2025] [Indexed: 05/11/2025] Open
Abstract
BACKGROUND Skin barrier dysfunction and immune activation are hallmarks of inflammatory skin diseases such as rosacea and psoriasis, suggesting shared pathogenic mechanisms. While barrier disruption may trigger or exacerbate skin inflammation, the precise underlying mechanisms remain unclear. Notably, epidermal barrier compromise leads to a marked increase in barrier alarmin expression. Among these, keratin 6A (KRT6A) plays a role in maintaining skin barrier integrity. METHODS We treated mouse skin and human keratinocytes, with and without KRT6A expression, with LL37/TNF-α and assessed the severity of inflammation. The specific mechanism by which KRT6A promotes skin inflammation was investigated using mass spectrometry and immunoprecipitation assays. RESULTS KRT6A expression was elevated in lesional skin from patients and mouse models of rosacea and psoriasis. In mice with LL37-induced rosacea-like and imiquimod (IMQ)-induced psoriasis-like skin inflammation, KRT6A knockdown alleviated inflammation, whereas KRT6A overexpression exacerbated inflammatory responses. Mechanistically, KRT6A activated signal transducer and activator of transcription 3 (STAT3) and enhanced proinflammatory cytokine expression in keratinocytes by reducing Janus kinase 1 (JAK1) ubiquitination. This occurred through inhibition of ring finger protein 41 (RNF41)-mediated JAK1 binding. CONCLUSIONS Our findings indicate that KRT6A expression increases following epidermal barrier disruption and contributes to exacerbated skin inflammation in disease conditions. Targeting KRT6A may represent a novel therapeutic approach for inflammatory skin diseases associated with epidermal dysfunction.
Collapse
Affiliation(s)
- Mengting Chen
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Aging Biology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Yaling Wang
- Department of Dermatology, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, China
| | - Mei Wang
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Aging Biology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - San Xu
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Aging Biology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Zixin Tan
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Aging Biology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Yisheng Cai
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Aging Biology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Xin Xiao
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Aging Biology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Ben Wang
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China.
- Hunan Key Laboratory of Aging Biology, Xiangya Hospital, Central South University, Changsha, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.
| | - Zhili Deng
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China.
- Hunan Key Laboratory of Aging Biology, Xiangya Hospital, Central South University, Changsha, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.
| | - Ji Li
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China.
- Hunan Key Laboratory of Aging Biology, Xiangya Hospital, Central South University, Changsha, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.
| |
Collapse
|
5
|
Fahmy LM, Adeuyan OO, de Jong A. CD1a and Lipids as Mediators of T Cell Activation in the Skin. J Invest Dermatol 2025:S0022-202X(25)00383-5. [PMID: 40317277 DOI: 10.1016/j.jid.2025.03.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 03/12/2025] [Accepted: 03/17/2025] [Indexed: 05/07/2025]
Abstract
Despite increasing insights in cytokine pathways involved in T cell-mediated inflammatory skin diseases, the upstream T cell triggering events through antigen-presenting molecules and antigens often remain incompletely understood. Recent studies have proposed an immunopathogenic role for T cells that are activated through CD1a, a lipid antigen-presenting molecule abundantly expressed on antigen-presenting cells in human skin. These CD1a-restricted T cells are thought to play a role in psoriasis, atopic dermatitis, and allergic contact dermatitis. In this review, we discuss modes of T cell activation by CD1a proteins and lipid antigens and bacteria as well as recent insights in local and systemic functions of CD1a-restricted T cells in inflammatory skin disease.
Collapse
Affiliation(s)
- Lauren M Fahmy
- Department of Dermatology, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, New York, USA
| | - Oluwaseyi O Adeuyan
- Department of Dermatology, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, New York, USA
| | - Annemieke de Jong
- Department of Dermatology, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, New York, USA.
| |
Collapse
|
6
|
Mima Y, Yamamoto M, Iozumi K. Cutaneous Adverse Events Following Nemolizumab Administration: A Review. J Clin Med 2025; 14:3026. [PMID: 40364058 PMCID: PMC12072469 DOI: 10.3390/jcm14093026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2025] [Revised: 04/25/2025] [Accepted: 04/25/2025] [Indexed: 05/15/2025] Open
Abstract
Atopic dermatitis (AD) is a chronic inflammatory skin disease characterized by epidermal barrier dysfunction and immune dysregulation, with interleukin (IL)-4, IL-13, and IL-31 recognized as key mediators. Prurigo nodularis (PN) is another chronic inflammatory disorder driven by T helper type 2-mediated inflammation and neural dysregulation, leading to severe pruritus. Nemolizumab, a humanized monoclonal antibody targeting IL-31 receptor A, has been approved for use in the treatment of AD and PN. Clinical trials have demonstrated significant reductions in pruritus and cutaneous symptoms associated with its use. In clinical practice, acute eczema and edematous erythema frequently occur, occasionally necessitating the discontinuation of treatment. Despite these observations, no comprehensive review has examined nemolizumab-associated cutaneous adverse events. This review aimed to examine various cutaneous reactions associated with nemolizumab therapy, including psoriasiform eruptions, AD exacerbation, bullous pemphigoid, drug-induced eruptions, and fungal infections. Potential mechanisms underlying these reactions include T-cell activation due to drug sensitization, immune responses triggered by nemolizumab acting as a hapten, and a relative increase in IL-4 and IL-13 levels following IL-31 inhibition. However, the precise pathophysiological mechanism and risk factors remain unclear, and standardized clinical management guidelines are lacking. Further accumulation of clinical data and immunological research are essential for developing evidence-based strategies to manage these adverse events, ensuring treatment continuity and optimizing patient outcomes.
Collapse
Affiliation(s)
- Yoshihito Mima
- Department of Dermatology, Tokyo Metropolitan Police Hospital, Tokyo 164-8541, Japan
| | | | | |
Collapse
|
7
|
Wang X, Mao D, Mu Z. IL-4, IL-13, TNF-α and IFN-γ Downregulate CLDN8 Expression Through Activating JAK Signaling Pathway in HaCaT Cells. Clin Cosmet Investig Dermatol 2025; 18:999-1009. [PMID: 40296865 PMCID: PMC12036616 DOI: 10.2147/ccid.s514527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Accepted: 04/15/2025] [Indexed: 04/30/2025]
Abstract
Background Tight junctions (TJs) are important for skin barrier function. Claudin-8 (CLDN8), a member of TJs, was indicated decreased in several RNA sequencing studies in dermatitis conditions. Methods Bioinformatics analysis was performed to extract CLDN8 mRNA expression from atopic dermatitis (AD) related datasets in the Gene Expression Omnibus. CLDN8 protein expression was detected in AD lesions and healthy control skin tissues using immunohistochemistry staining (IHC). Cldn8 expression was detected in MC903-induced AD-like mouse model. AD-related cytokines with or without Janus kinase (JAK) inhibitor were added to HaCaT cells, and CLDN8 expression was detected by quantitative Polymerase Chain Reaction (qPCR). Results CLDN8 mRNA expression is decreased in AD lesions and MC903-induced AD-like mouse model. Downregulation of CLDN8 mRNA expression is alleviated after dupilumab or crisaborole treatment. CLDN8 protein was not detected by IHC in human or mouse skin tissues. Interleukin (IL)-4, IL-13, tumor necrosis factor (TNF)-α and interferon (IFN)-γ downregulated CLDN8 mRNA expression in HaCaT cells through activating JAK. Conclusion CLDN8 mRNA is decreased in AD lesions, and the decreased CLDN8 is alleviated along with therapy. Skin tissues might not express CLDN8 protein. AD-related cytokines including IL-4, IL-13, TNF-α and IFN-γ could downregulate CLDN8 mRNA expression through activating JAK.
Collapse
Affiliation(s)
- Xiaojie Wang
- Department of Dermatology, Peking University People’s Hospital, Beijing, People’s Republic of China
| | - Dandan Mao
- Department of Dermatology, Peking University People’s Hospital, Beijing, People’s Republic of China
| | - Zhanglei Mu
- Department of Dermatology, Peking University People’s Hospital, Beijing, People’s Republic of China
| |
Collapse
|
8
|
Liu Z, Guo M, Li Y, Xu H. A multidimensional analysis of prognostic factors in atopic dermatitis. Front Med (Lausanne) 2025; 12:1554669. [PMID: 40313541 PMCID: PMC12043433 DOI: 10.3389/fmed.2025.1554669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Accepted: 03/31/2025] [Indexed: 05/03/2025] Open
Abstract
Atopic dermatitis (AD) is a chronic inflammatory skin condition with a high prevalence worldwide and multifaceted pathogenesis. In general, patients with moderate to severe AD often experience relapse after discontinuing treatment. Therefore, to understand the possible factors of chronic relapse of AD and to look for biological markers that predict the relapse or poor prognosis of AD will be helpful for clinical treatment. Mutations in genes such as FLG, SPINK5, STAT, KIF3A, claudin-1, Ovol1, and HLA-DRB1 offer new insights into the genetic basis of AD. Routine factors may help improve patient lifestyle, highlight the importance of environmental influences (including psychological stress), and support clinicians in optimizing anti-infective treatment strategies. The inflammatory axis (CD30-CD30L axis, IL-9-IL-18 axis) provides new insights into the inflammatory pathways of AD and may be a target for future therapies. Low NKG2D expression may have adverse effects on prognosis. Prognostic biomarkers can play an important role in treatment monitoring, disease progression and recurrence, and provide the possibility for more personalized treatment.
Collapse
Affiliation(s)
| | | | | | - Hui Xu
- Department of Dermatology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| |
Collapse
|
9
|
Blicharz L, Michalczyk A, Maj M, Czuwara J, Olszewska M, Rudnicka L. Head and neck dermatitis: a variant of atopic dermatitis. Ital J Dermatol Venerol 2025; 160:123-144. [PMID: 40248964 DOI: 10.23736/s2784-8671.25.08001-6] [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: 04/19/2025]
Abstract
Atopic dermatitis involves the head and neck area across all age groups. This manifestation is frequently referred to as "the head and neck dermatitis." Aside from a considerable deterioration of the quality of life, it poses a significant diagnostic and therapeutic challenge. The head and neck dermatitis may be mimicked by other inflammatory conditions such as seborrheic dermatitis or contact dermatitis. Furthermore, it can be associated with a wide range of infectious, ocular, psychiatric and hair disorders, which should raise clinical alertness and encourage a multidisciplinary management of the affected individuals. Skin lesions in the head and neck area are often difficult to treat, particularly because of a considerable exposure of this region to exacerbating factors and limitations regarding the use of some pharmaceuticals. Although several hypotheses explaining the recalcitrant course of head and neck dermatitis have been proposed, none of them provide successful solutions applicable in the daily clinical practice. This comprehensive review comprises the current insights on the pathogenesis, clinical presentation, and comorbidities of the head and neck dermatitis. Recommendations regarding possible treatments of this condition such as antifungals, as well as special considerations for the choice of biologics or JAK inhibitors in the candidates for systemic treatment are outlined.
Collapse
Affiliation(s)
- Leszek Blicharz
- Department of Dermatology, Medical University of Warsaw, Warsaw, Poland
| | | | - Małgorzata Maj
- Department of Dermatology, Medical University of Warsaw, Warsaw, Poland
| | - Joanna Czuwara
- Department of Dermatology, Medical University of Warsaw, Warsaw, Poland -
| | | | - Lidia Rudnicka
- Department of Dermatology, Medical University of Warsaw, Warsaw, Poland
| |
Collapse
|
10
|
Saeki H, Ohya Y, Arakawa H, Ichiyama S, Katsunuma T, Katoh N, Tanaka A, Tanizaki H, Tsunemi Y, Nakahara T, Nagao M, Narita M, Hide M, Fujisawa T, Futamura M, Masuda K, Matsubara T, Murota H, Yamamoto-Hanada K, Furuta J. Executive summary: Japanese guidelines for atopic dermatitis (ADGL) 2024. Allergol Int 2025; 74:210-221. [PMID: 39986987 DOI: 10.1016/j.alit.2025.01.003] [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: 12/24/2024] [Accepted: 01/23/2025] [Indexed: 02/24/2025] Open
Abstract
This is an abridged edition of English version of the Clinical Practice Guidelines for the Management of Atopic Dermatitis 2024. Atopic dermatitis (AD) is a disease characterized by relapsing eczema with pruritus as a primary lesion. A crucial aspect of AD treatment is the prompt induction of remission via the suppression of existing skin inflammation and pruritus. To achieve this, topical anti-inflammatory drugs such as topical corticosteroids, tacrolimus ointment, delgocitinib ointment, and difamilast ointment have been used. However, the following treatments should be considered in addition to topical therapy for patients with refractory moderate-to-severe AD: oral cyclosporine, subcutaneous injections of biologics (dupilumab, nemolizumab, tralokinumab), oral Janus kinase inhibitors (baricitinib, upadacitinib, abrocitinib), and phototherapy. In the revised guidelines, descriptions of five new drugs, namely, difamilast, nemolizumab, tralokinumab, upadacitinib, and abrocitinib, have been added. The guidelines present recommendations to review clinical research articles, evaluate the balance between the advantages and disadvantages of medical activities, and optimize medical activity-related patient outcomes with respect to several important points requiring decision-making in clinical practice.
Collapse
Affiliation(s)
- Hidehisa Saeki
- Department of Dermatology, Nippon Medical School, Tokyo, Japan.
| | - Yukihiro Ohya
- Department of Occupational and Environmental Health, Graduate School of Medical Sciences and Medical School, Nagoya City University, Nagoya, Japan
| | - Hirokazu Arakawa
- Kitakanto Allergy Research Institute, Kibounoie Hospital, Gunma, Japan
| | - Susumu Ichiyama
- Department of Dermatology, Nippon Medical School, Tokyo, Japan
| | - Toshio Katsunuma
- Department of Pediatrics, The Jikei University Daisan Hospital, Tokyo, Japan
| | - Norito Katoh
- North Campus of Kyoto Prefectural University of Medicine and Department for Medical Innovation and Translational Medical Science, Kyoto Prefectural University of Medicine Graduate School of Medical Science, Kyoto, Japan
| | - Akio Tanaka
- Department of Dermatology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Hideaki Tanizaki
- Department of Dermatology, Kansai Medical University, Osaka, Japan
| | - Yuichiro Tsunemi
- Department of Dermatology, Saitama Medical University, Saitama, Japan
| | - Takeshi Nakahara
- Department of Dermatology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Mizuho Nagao
- Allergy Center, National Hospital Organization Mie National Hospital, Tsu, Japan
| | - Masami Narita
- Department of Pediatrics, Faculty of Medicine, Kyorin University, Tokyo, Japan
| | - Michihiro Hide
- Department of Dermatology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan; Department of Dermatology, Hiroshima City Hiroshima Citizens Hospital, Hiroshima, Japan
| | - Takao Fujisawa
- Allergy Center, National Hospital Organization Mie National Hospital, Tsu, Japan
| | - Masaki Futamura
- Division of Pediatrics, National Hospital Organization Nagoya Medical Center, Nagoya, Japan
| | - Koji Masuda
- Department of Dermatology, Kyoto Prefectural University of Medicine Graduate School of Medical Science, Kyoto, Japan
| | - Tomoyo Matsubara
- Department of Pediatrics, Dokkyo Medical University Saitama Medical Center, Saitama, Japan
| | - Hiroyuki Murota
- Department of Dermatology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | | | - Junichi Furuta
- Medical Informatics and Management, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| |
Collapse
|
11
|
Díez-Madueño K, Montero I, Fernández-Gosende M, Martínez-Álvarez N, Hidalgo-Cantabrana C, de la Cueva Dobao P, Coto-Segura P. Compositional and Functional Profile of Gut Microbiota in a Cohort of Adult Spanish Patients with Atopic Dermatitis Using Metagenomics: A Cross-Sectional Study. Dermatitis 2025. [PMID: 40111891 DOI: 10.1089/derm.2024.0536] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/22/2025]
Abstract
Background: The role of gut dysbiosis in the pathophysiology of atopic dermatitis (AD) through immune system (IS) imbalance is a novel line of investigation currently under discussion. This study aimed to characterize compare the composition and functional profile of the gut microbiota (GM) between adults with AD and healthy individuals. Methods: Observational cross-sectional study, where fecal samples from 70 adults (38 patients and 32 controls) were analyzed using metagenomics and bioinformatics. Results: Differences between the GM of patients with AD and healthy individuals were demonstrated. Reduced microbial diversity was found in subjects with AD. Bacterial species with lower abundance primarily belonged to the families Ruminococcaceae, Akkermansiaceae, and Methanobacteriaceae. Several microbial metabolic pathways were found to be decreased in patients with AD, including amino acid biosynthesis, vitamin biosynthesis, fatty acids and lipids biosynthesis, and energy metabolism. Conclusion: Adults with AD exhibited a distinct GM compared to healthy individuals. Changes were demonstrated both compositionally and functionally. Further investigation is mandatory to elucidate the potential link and causal relationship between gut dysbiosis and AD, which may be crucial for a deeper understanding of the disease's pathophysiology and the development of novel therapeutic approaches.
Collapse
Affiliation(s)
- Kevin Díez-Madueño
- From the Dermatology Department, Hospital Universitario Infanta Leonor, Madrid, Spain
- Complutense University of Madrid, Madrid, Spain
| | | | | | | | | | - Pablo de la Cueva Dobao
- From the Dermatology Department, Hospital Universitario Infanta Leonor, Madrid, Spain
- Complutense University of Madrid, Madrid, Spain
| | - Pablo Coto-Segura
- Dermatology Department, Hospital Vital Álvarez Buylla, Mieres, Spain
| |
Collapse
|
12
|
Ullah S, Feng F, Zhao M, Zhang J, Shao Q. Comparative Effects of Dietary Supplementations with Microencapsulated Sodium Butyrate, Glycerol Monolaurate and Tributyrin on Growth, Immunity, and Gut Health in Black Sea Bream. Animals (Basel) 2025; 15:810. [PMID: 40150339 PMCID: PMC11939239 DOI: 10.3390/ani15060810] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2025] [Revised: 03/07/2025] [Accepted: 03/10/2025] [Indexed: 03/29/2025] Open
Abstract
This study investigates the effects of three dietary additives-microencapsulated sodium butyrate (MSB), glycerol monolaurate (GML), and tributyrin (TB)-on the growth performance, various physiological parameters, gene expression, intestinal morphology, and microflora in Acanthopagrus schlegelii (black sea bream). The experiment utilized a 43.5% soybean meal (SBM) inclusion diet with four isonitrogenous and isoenergetic formulations: a control diet, and diets supplemented with MSB (0.24%), GML (0.04%), or TB (0.22%). The growth trial spanned eight weeks, and triplicate tanks were randomly assigned to each diet, with each tank containing 30 fish, each having an initial weight of 1.55 ± 0.01 g. Key outcomes included measurements of weight gain, specific growth rate, digestive enzyme activity, serum immune markers, antioxidant status, and intestinal morphology and, gut microbiota. Additionally, gene expression and microbiota analysis were conducted on intestinal tissues to assess the impact of these additives on gut health and immune response. The findings revealed that all three additives enhanced growth performance and improved intestinal health and gut microbiota but GML exhibited the most pronounced effects on intestinal barrier function and immune modulation, gene expression, and microflora, followed by MSB and TB. This study provides a comprehensive comparison of MSB, GML, and TB as feed additives for black sea bream, offering insights into their potential for improving fish health and optimizing aquaculture feed formulations.
Collapse
Affiliation(s)
- Sami Ullah
- Zhejiang University Zhongyuan Institute, Zhengzhou 450001, China; (S.U.); (F.F.)
- College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou 310058, China;
| | - Fengqin Feng
- Zhejiang University Zhongyuan Institute, Zhengzhou 450001, China; (S.U.); (F.F.)
- College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou 310058, China;
| | - Minjie Zhao
- College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou 310058, China;
| | - Jinzhi Zhang
- College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| | - Qingjun Shao
- College of Animal Sciences, Zhejiang University, Hangzhou 310058, China
| |
Collapse
|
13
|
Jaros J, Ahuja K, Lio P. Exploring the Link Between Atopic Dermatitis and Eosinophilic Esophagitis. THE JOURNAL OF CLINICAL AND AESTHETIC DERMATOLOGY 2025; 18:15-20. [PMID: 40135176 PMCID: PMC11932097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 03/27/2025]
Abstract
Eosinophilic esophagitis (EoE) and atopic dermatitis (AD) are two known and sometimes comorbid type 2 helper cell-mediated diseases. EoE shares clinical features, immunologic pathways, susceptibility loci, and risk with atopic conditions including food allergies (food allergies), asthma, allergic rhinitis (AR), and AD. These conditions share an impaired immunological response against a range of antigens or allergens, leading to CD4+ Th2 differentiation and overproduction of immunoglobulin E (IgE). The emerging coexistence of EoE and AD presents a compelling area of study. Both diseases manifest on stratified squamous epithelium along the skin-gut continuum and have overlapping treatment algorithms that include avoidance of triggers, topical steroids, and dupilumab. This narrative review highlights the clinical and immunologic nuances underlying these two conditions and sheds light on potential new research and therapeutic avenues.
Collapse
Affiliation(s)
- Joanna Jaros
- Dr. Jaros is with the Department of Dermatology at Cook County Hospital and Health System in Chicago, Illinois
| | - Kripa Ahuja
- Ms. Ahuja is with Eastern Virginia Medical School in Norfolk, Virginia
| | - Peter Lio
- Dr. Lio is with the Department of Dermatology at Northwestern University in Chicago, Illinois
| |
Collapse
|
14
|
Combarros D, Brahmi R, Musaefendic E, Heit A, Kondratjeva J, Moog F, Pressanti C, Lecru LA, Arbouille S, Laffort C, Goudounèche D, Brun J, Simon M, Cadiergues MC. Reconstructed Epidermis Produced with Atopic Dog Keratinocytes Only Exhibit Skin Barrier Defects after the Addition of Proinflammatory and Allergic Cytokines. JID INNOVATIONS 2025; 5:100330. [PMID: 39811760 PMCID: PMC11730559 DOI: 10.1016/j.xjidi.2024.100330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 10/29/2024] [Accepted: 11/05/2024] [Indexed: 01/16/2025] Open
Abstract
Our objectives were to explore epidermal barrier defects in dogs with atopic dermatitis and to determine whether the defects are genetically determined or secondary to skin inflammation. First, the expression of filaggrin, corneodesmosin, and claudin1, analyzed using indirect immunofluorescence in skin biopsies collected from 32 healthy and 32 dogs with atopic dermatitis, was weaker in the atopic skin (P = .003). Second, primary keratinocytes of atopic dogs and healthy dogs were used to produce 3-dimensional reconstructed canine epidermis. The expression of the same proteins was analyzed using indirect immunofluorescence, immunoblotting, and RT-qPCR, whereas reconstructed canine epidermis morphology was investigated by transmission electron microscopy, and the barrier was investigated by functional assays. Next, inflammatory cytokines (IL-4, IL-13, IL-31, and TNFα) were added to the culture medium. The morphology, protein expression, and barrier function of the reconstructed canine epidermis were similar whether produced with keratinocytes from healthy dogs or dogs with atopy. Addition of inflammatory cytokines impaired the protein expression and epidermal barrier of the 2 types of reconstructed canine epidermis equally. To conclude, the reduced expression of epidermal barrier proteins observed in vivo was not reproduced in vitro unless cytokines were used, suggesting that it is induced by the inflammatory milieu.
Collapse
Affiliation(s)
- Daniel Combarros
- Small Animal Clinic, École Nationale Vétérinaire de Toulouse, University of Toulouse, Toulouse, France
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity), University of Toulouse, INSERM, CNRS, Paul Sabatier Toulouse III University, Toulouse, France
| | - Rahma Brahmi
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity), University of Toulouse, INSERM, CNRS, Paul Sabatier Toulouse III University, Toulouse, France
| | - Emma Musaefendic
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity), University of Toulouse, INSERM, CNRS, Paul Sabatier Toulouse III University, Toulouse, France
| | - Alizée Heit
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity), University of Toulouse, INSERM, CNRS, Paul Sabatier Toulouse III University, Toulouse, France
| | - Jevgenija Kondratjeva
- Small Animal Clinic, École Nationale Vétérinaire de Toulouse, University of Toulouse, Toulouse, France
| | - Fabien Moog
- Small Animal Clinic, École Nationale Vétérinaire de Toulouse, University of Toulouse, Toulouse, France
| | - Charline Pressanti
- Small Animal Clinic, École Nationale Vétérinaire de Toulouse, University of Toulouse, Toulouse, France
| | - Line A. Lecru
- Clinique vétérinaire Hermes-Plage, Marseille, France
| | | | | | - Dominique Goudounèche
- Centre de Microscopie Electronique Appliquée à la Biologie, University of Toulouse, Toulouse, France
| | - Jessie Brun
- Small Animal Clinic, École Nationale Vétérinaire de Toulouse, University of Toulouse, Toulouse, France
| | - Michel Simon
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity), University of Toulouse, INSERM, CNRS, Paul Sabatier Toulouse III University, Toulouse, France
| | - Marie-Christine Cadiergues
- Small Animal Clinic, École Nationale Vétérinaire de Toulouse, University of Toulouse, Toulouse, France
- Toulouse Institute for Infectious and Inflammatory Diseases (Infinity), University of Toulouse, INSERM, CNRS, Paul Sabatier Toulouse III University, Toulouse, France
| |
Collapse
|
15
|
Ogulur I, Mitamura Y, Yazici D, Pat Y, Ardicli S, Li M, D'Avino P, Beha C, Babayev H, Zhao B, Zeyneloglu C, Giannelli Viscardi O, Ardicli O, Kiykim A, Garcia-Sanchez A, Lopez JF, Shi LL, Yang M, Schneider SR, Skolnick S, Dhir R, Radzikowska U, Kulkarni AJ, Imam MB, Veen WVD, Sokolowska M, Martin-Fontecha M, Palomares O, Nadeau KC, Akdis M, Akdis CA. Type 2 immunity in allergic diseases. Cell Mol Immunol 2025; 22:211-242. [PMID: 39962262 PMCID: PMC11868591 DOI: 10.1038/s41423-025-01261-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Accepted: 01/09/2025] [Indexed: 03/01/2025] Open
Abstract
Significant advancements have been made in understanding the cellular and molecular mechanisms of type 2 immunity in allergic diseases such as asthma, allergic rhinitis, chronic rhinosinusitis, eosinophilic esophagitis (EoE), food and drug allergies, and atopic dermatitis (AD). Type 2 immunity has evolved to protect against parasitic diseases and toxins, plays a role in the expulsion of parasites and larvae from inner tissues to the lumen and outside the body, maintains microbe-rich skin and mucosal epithelial barriers and counterbalances the type 1 immune response and its destructive effects. During the development of a type 2 immune response, an innate immune response initiates starting from epithelial cells and innate lymphoid cells (ILCs), including dendritic cells and macrophages, and translates to adaptive T and B-cell immunity, particularly IgE antibody production. Eosinophils, mast cells and basophils have effects on effector functions. Cytokines from ILC2s and CD4+ helper type 2 (Th2) cells, CD8 + T cells, and NK-T cells, along with myeloid cells, including IL-4, IL-5, IL-9, and IL-13, initiate and sustain allergic inflammation via T cell cells, eosinophils, and ILC2s; promote IgE class switching; and open the epithelial barrier. Epithelial cell activation, alarmin release and barrier dysfunction are key in the development of not only allergic diseases but also many other systemic diseases. Recent biologics targeting the pathways and effector functions of IL4/IL13, IL-5, and IgE have shown promising results for almost all ages, although some patients with severe allergic diseases do not respond to these therapies, highlighting the unmet need for a more detailed and personalized approach.
Collapse
Affiliation(s)
- Ismail Ogulur
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Yasutaka Mitamura
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Duygu Yazici
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Yagiz Pat
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Sena Ardicli
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
- Department of Genetics, Faculty of Veterinary Medicine, Bursa Uludag University, Bursa, Turkey
| | - Manru Li
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Paolo D'Avino
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Carina Beha
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Huseyn Babayev
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Bingjie Zhao
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Can Zeyneloglu
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | | | - Ozge Ardicli
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
- Division of Food Processing, Milk and Dairy Products Technology Program, Karacabey Vocational School, Bursa Uludag University, Bursa, Turkey
| | - Ayca Kiykim
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
- Department of Pediatrics, Division of Pediatric Allergy and Immunology, Cerrahpasa School of Medicine, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Asuncion Garcia-Sanchez
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
- Department of Biomedical and Diagnostic Science, School of Medicine, University of Salamanca, Salamanca, Spain
| | - Juan-Felipe Lopez
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Li-Li Shi
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
- Department of Otolaryngology-Head and Neck Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Minglin Yang
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Stephan R Schneider
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Stephen Skolnick
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
- Seed Health Inc., Los Angeles, CA, USA
| | - Raja Dhir
- Seed Health Inc., Los Angeles, CA, USA
| | - Urszula Radzikowska
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Abhijeet J Kulkarni
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Manal Bel Imam
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Willem van de Veen
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Milena Sokolowska
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Mar Martin-Fontecha
- Departamento de Quimica Organica, Facultad de Optica y Optometria, Complutense University of Madrid, Madrid, Spain
| | - Oscar Palomares
- Department of Biochemistry and Molecular Biology, School of Chemistry, Complutense University of Madrid, Madrid, Spain
| | - Kari C Nadeau
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Mubeccel Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Cezmi A Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland.
| |
Collapse
|
16
|
Ramirez GA, Cardamone C, Lettieri S, Fredi M, Mormile I. Clinical and Pathophysiological Tangles Between Allergy and Autoimmunity: Deconstructing an Old Dichotomic Paradigm. Clin Rev Allergy Immunol 2025; 68:13. [PMID: 39932658 PMCID: PMC11814061 DOI: 10.1007/s12016-024-09020-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/26/2024] [Indexed: 02/14/2025]
Abstract
Allergic and autoimmune disorders are characterised by dysregulation of the immune responses to otherwise inert environmental substances and autoantigens, leading to inflammation and tissue damage. Their incidence has constantly increased in the last decades, and their co-occurrence defies current standards in patient care. For years, allergy and autoimmunity have been considered opposite conditions, with IgE and Th2 lymphocytes cascade driving canonical allergic manifestations and Th1/Th17-related pathways accounting for autoimmunity. Conversely, growing evidence suggests that these conditions not only share some common inciting triggers but also are subtended by overlapping pathogenic pathways. Permissive genetic backgrounds, along with epithelial barrier damage and changes in the microbiome, are now appreciated as common risk factors for both allergy and autoimmunity. Eosinophils and mast cells, along with autoreactive IgE, are emerging players in triggering and sustaining autoimmunity, while pharmacological modulation of B cells and Th17 responses has provided novel clues to the pathophysiology of allergy. By combining clinical and therapeutic evidence with data from mechanistic studies, this review provides a state-of-the-art update on the complex interplay between allergy and autoimmunity, deconstructing old dichotomic paradigms and offering potential clues for future research.
Collapse
Affiliation(s)
- Giuseppe A Ramirez
- Unit of Immunology, Rheumatology, Allergy and Rare Diseases, IRCCS Ospedale San Raffaele, Milan, Italy
- Università Vita-Salute San Raffaele, Milan, Italy
| | - Chiara Cardamone
- Immunorheumatology Unit, University Hospital "San Giovanni Di Dio E Ruggi d'Aragona", Largo Città d'Ippocrate, Via San Leonardo 1, 84131, Salerno, Italy.
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, Baronissi, Italy.
| | - Sara Lettieri
- Pulmonology Unit, IRCCS San Matteo Hospital Foundation, Pavia, Italy
- Department of Internal Medicine and Therapeutics, University of Pavia, Pavia, Italy
| | - Micaela Fredi
- Rheumatology and Clinical Immunology Unit, ASST Spedali Civili of Brescia, Brescia, Italy
- Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Ilaria Mormile
- Division of Internal Medicine and Clinical Immunology, Department of Internal Medicine and Clinical Complexity, AOU Federico II, Naples, Italy
- Department of Translational Medical Sciences, Federico II University, Naples, Italy
| |
Collapse
|
17
|
Zhang J, Li G, Guo Q, Yang Y, Yang J, Feng X, Yao Z. Allergens in Atopic Dermatitis. Clin Rev Allergy Immunol 2025; 68:11. [PMID: 39924626 DOI: 10.1007/s12016-025-09024-7] [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] [Accepted: 01/20/2025] [Indexed: 02/11/2025]
Abstract
Atopic dermatitis (AD) is a chronic inflammatory skin disease with a complex relationship to allergens. While AD itself is not an allergic reaction and does not necessarily involve allergen sensitization, AD patients show higher rates of sensitization to food and inhalant allergens compared to the general population. Recent evidence refining the "dual allergen exposure hypothesis" demonstrates that early oral exposure to allergens through an intact gastrointestinal barrier typically promotes tolerance, while exposure through compromised skin or respiratory barriers often leads to sensitization. Therefore, the impaired skin barrier function in AD patients increases the risk of transcutaneous sensitization and may interfere with oral tolerance development. Interestingly, AD patients' sensitivity to contact allergens (such as metals and fragrances) is not necessarily higher than that of the general population, which may be related to the inherent properties of these allergens. Personalized allergen testing can help guide appropriate allergen avoidance and reintroduction strategies in AD management. The insights into optimal allergen exposure conditions have also expanded the potential applications of allergen-specific immunotherapy in preventing AD onset in high-risk populations and halting the atopic march.
Collapse
Affiliation(s)
- Jiayan Zhang
- Dermatology Center, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, 1665 Kongjiang Road, Shanghai, 200092, China
- Department of Dermatology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, 1665 Kongjiang Road, Shanghai, 200092, China
- Institute of Dermatology, Shanghai Jiaotong University School of Medicine, 1665 Kongjiang Road, Shanghai, 200092, China
| | - Guofang Li
- Dermatology Center, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, 1665 Kongjiang Road, Shanghai, 200092, China
- Department of Dermatology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, 1665 Kongjiang Road, Shanghai, 200092, China
- Institute of Dermatology, Shanghai Jiaotong University School of Medicine, 1665 Kongjiang Road, Shanghai, 200092, China
| | - Qiuyang Guo
- Dermatology Center, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, 1665 Kongjiang Road, Shanghai, 200092, China
- Department of Dermatology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, 1665 Kongjiang Road, Shanghai, 200092, China
- Institute of Dermatology, Shanghai Jiaotong University School of Medicine, 1665 Kongjiang Road, Shanghai, 200092, China
| | - Yijun Yang
- Dermatology Center, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, 1665 Kongjiang Road, Shanghai, 200092, China
- Department of Dermatology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, 1665 Kongjiang Road, Shanghai, 200092, China
- Institute of Dermatology, Shanghai Jiaotong University School of Medicine, 1665 Kongjiang Road, Shanghai, 200092, China
| | - Jinxiang Yang
- Dermatology Center, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, 1665 Kongjiang Road, Shanghai, 200092, China
- Department of Dermatology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, 1665 Kongjiang Road, Shanghai, 200092, China
- Institute of Dermatology, Shanghai Jiaotong University School of Medicine, 1665 Kongjiang Road, Shanghai, 200092, China
| | - Xiaobo Feng
- Dermatology Center, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, 1665 Kongjiang Road, Shanghai, 200092, China.
- Department of Dermatology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, 1665 Kongjiang Road, Shanghai, 200092, China.
- Institute of Dermatology, Shanghai Jiaotong University School of Medicine, 1665 Kongjiang Road, Shanghai, 200092, China.
| | - Zhirong Yao
- Dermatology Center, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, 1665 Kongjiang Road, Shanghai, 200092, China.
- Department of Dermatology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, 1665 Kongjiang Road, Shanghai, 200092, China.
- Institute of Dermatology, Shanghai Jiaotong University School of Medicine, 1665 Kongjiang Road, Shanghai, 200092, China.
| |
Collapse
|
18
|
Saeki H, Ohya Y, Arakawa H, Ichiyama S, Katsunuma T, Katoh N, Tanaka A, Tanizaki H, Tsunemi Y, Nakahara T, Nagao M, Narita M, Hide M, Fujisawa T, Futamura M, Masuda K, Matsubara T, Murota H, Yamamoto-Hanada K, Furuta J. English version of clinical practice guidelines for the management of atopic dermatitis 2024. J Dermatol 2025; 52:e70-e142. [PMID: 39707640 DOI: 10.1111/1346-8138.17544] [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: 10/10/2024] [Accepted: 10/14/2024] [Indexed: 12/23/2024]
Abstract
This is the English version of the 2024 clinical practice guidelines for the management of atopic dermatitis (AD). AD is a disease characterized by relapsing eczema with pruritus as a primary lesion. A crucial aspect of AD treatment is the prompt induction of remission via the suppression of existing skin inflammation and pruritus. To achieve this, topical anti-inflammatory drugs, such as topical corticosteroids, tacrolimus ointment, delgocitinib ointment, and difamilast ointment, have been used. However, the following treatments should be considered in addition to topical therapy for patients with refractory moderate-to-severe AD: oral cyclosporine, subcutaneous injections of biologics (dupilumab, nemolizumab, tralokinumab), oral Janus kinase inhibitors (baricitinib, upadacitinib, abrocitinib), and phototherapy. In these revised guidelines, descriptions of five new drugs, namely, difamilast, nemolizumab, tralokinumab, upadacitinib, and abrocitinib, have been added. The guidelines present recommendations to review clinical research articles, evaluate the balance between the advantages and disadvantages of medical activities, and optimize medical activity-related patient outcomes with respect to several important points requiring decision-making in clinical practice.
Collapse
Affiliation(s)
- Hidehisa Saeki
- Department of Dermatology, Nippon Medical School, Tokyo, Japan
| | - Yukihiro Ohya
- Department of Occupational and Environmental Health, Graduate School of Medical Sciences and Medical School, Nagoya City University, Nagoya, Japan
| | - Hirokazu Arakawa
- Kitakanto Allergy Research Institute, Kibounoie Hospital, Gunma, Japan
| | - Susumu Ichiyama
- Department of Dermatology, Nippon Medical School, Tokyo, Japan
| | - Toshio Katsunuma
- Department of Pediatrics, The Jikei University Daisan Hospital, Tokyo, Japan
| | - Norito Katoh
- Department for Medical Innovation and Translational Medical Science, Kyoto Prefectural University of Medicine Graduate School of Medical Science, Kyoto, Japan
| | - Akio Tanaka
- Department of Dermatology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Hideaki Tanizaki
- Department of Dermatology, Kansai Medical University, Osaka, Japan
| | - Yuichiro Tsunemi
- Department of Dermatology, Saitama Medical University, Saitama, Japan
| | - Takeshi Nakahara
- Department of Dermatology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Mizuho Nagao
- Allergy Center, National Hospital Organization Mie National Hospital, Tsu, Japan
| | - Masami Narita
- Department of Pediatrics, Faculty of Medicine, Kyorin University, Tokyo, Japan
| | - Michihiro Hide
- Department of Dermatology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
- Department of Dermatology, Hiroshima City Hiroshima Citizens Hospital, Hiroshima, Japan
| | - Takao Fujisawa
- Allergy Center, National Hospital Organization Mie National Hospital, Tsu, Japan
| | - Masaki Futamura
- Division of Pediatrics, National Hospital Organization Nagoya Medical Center, Nagoya, Japan
| | - Koji Masuda
- Department of Dermatology, Kyoto Prefectural University of Medicine Graduate School of Medical Science, Kyoto, Japan
| | - Tomoyo Matsubara
- Department of Pediatrics, Dokkyo Medical University Saitama Medical Center, Saitama, Japan
| | - Hiroyuki Murota
- Department of Dermatology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | | | - Junichi Furuta
- Medical Informatics and Management, Institute of Medicine, University of Tsukuba, Tsukuba, Japan
| |
Collapse
|
19
|
Pan Y, Hochgerner M, Cichoń MA, Benezeder T, Bieber T, Wolf P. Langerhans cells: Central players in the pathophysiology of atopic dermatitis. J Eur Acad Dermatol Venereol 2025; 39:278-289. [PMID: 39157943 PMCID: PMC11760705 DOI: 10.1111/jdv.20291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Accepted: 07/21/2024] [Indexed: 08/20/2024]
Abstract
Atopic dermatitis (AD) is the most common chronic inflammatory skin disease worldwide. AD is a highly complex disease with different subtypes. Many elements of AD pathophysiology have been described, but if/how they interact with each other or which mechanisms are important in which patients is still unclear. Langerhans cells (LCs) are antigen-presenting cells (APCs) in the epidermis. Depending on the context, they can act either pro- or anti-inflammatory. Many different studies have investigated LCs in the context of AD and found them to be connected to all major mechanisms of AD pathophysiology. As APCs, LCs recruit other immune cells and shape the immune response, especially adaptive immunity via polarization of T cells. As sentinel cells, LCs are primary sensors of the skin microbiome and are important for the decision of immunity versus tolerance. LCs are also involved with the integrity of the skin barrier by influencing tight junctions. Finally, LCs are important cells in the neuro-immune crosstalk in the skin. In this review, we provide an overview about the many different roles of LCs in AD. Understanding LCs might bring us closer to a more complete understanding of this highly complex disease. Potentially, modulating LCs might offer new options for targeted therapies for AD patients.
Collapse
Affiliation(s)
- Yi Pan
- Department of Dermatology and AllergyUniversity Hospital of BonnBonnGermany
- Department of Dermatology and VenerologyMedical University of GrazGrazAustria
| | - Mathias Hochgerner
- Greater Bay Area Institute of Precision Medicine (Guangzhou), School of Life Sciences, Fudan UniversityShanghaiChina
| | | | - Theresa Benezeder
- Department of Dermatology and VenerologyMedical University of GrazGrazAustria
| | - Thomas Bieber
- Department of Dermatology and AllergyUniversity Hospital of BonnBonnGermany
- CK‐CARE, Medicine CampusDavosSwitzerland
- Department of DermatologyUniversity Hospital of ZürichZürichSwitzerland
| | - Peter Wolf
- Department of Dermatology and VenerologyMedical University of GrazGrazAustria
| |
Collapse
|
20
|
Sung YY, Yuk HJ, Kim SH, Kim DS. Effects of Earthworm ( Pheretima communisima) extract on atopic dermatitis: An in vitro and in vivo study. Heliyon 2025; 11:e41140. [PMID: 39758409 PMCID: PMC11699427 DOI: 10.1016/j.heliyon.2024.e41140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 12/05/2024] [Accepted: 12/10/2024] [Indexed: 01/07/2025] Open
Abstract
Earthworm (Pheretima communisima) is used as a traditional medicine for the management of allergic airway inflammation. Atopic dermatitis (AD) is a persistent, recurrent disorder marked by allergic inflammation and skin barrier dysfunction. However, the pharmaceutical effects of earthworms on AD have not been defined. Our study examined the anti-allergic and anti-inflammatory actions of earthworm ethanolic extract (EWE) on allergic skin inflammation in a Dermatophagoides farinae mite antigen-induced AD mice model, TNF-α/IFN-γ-treated human keratinocytes, and compound 48/80-treated mouse mast cells. Oral administration of EWE in AD mouse reduced inflammatory cell accumulation, epidermal hyperplasia, and dermatitis severity in AD skin lesions and thymic stromal lymphopoietin (TSLP) and immunoglobulin (Ig) E concentrations in serum. EWE administration in AD mice also reduced secretion of Interleukin (IL)-4, IL-13, IL-5, and IFN-γ in cultures of isolated splenic cells. Immunohistofluorescence staining of skin lesions from AD mice revealed that EWE induced expression of claudin-1, filaggrin, and SIRT1. In HaCaT keratinocytes cotreated with IFN-γ and TNF-α, EWE inhibited secretion of the chemokine Regulated on Activation, Normal T Cell Expressed and Secreted (RANTES) in a dose-dependent manner. In addition, EWE inhibited histamine release in activated MC/9 mast cells. These results show that EWE might be therapeutics for the management of AD.
Collapse
Affiliation(s)
- Yoon-Young Sung
- KM Science Research Division, Korea Institute of Oriental Medicine, 1672 Yuseongdae-ro, Yuseong-gu, Daejeon, 34054, South Korea
| | - Heung-Joo Yuk
- KM Science Research Division, Korea Institute of Oriental Medicine, 1672 Yuseongdae-ro, Yuseong-gu, Daejeon, 34054, South Korea
| | - Seung-Hyung Kim
- Institute of Traditional Medicine and Bioscience, Daejeon University, 62 Daehak-ro, Dong-gu, Daejeon, 34520, South Korea
| | - Dong-Seon Kim
- KM Science Research Division, Korea Institute of Oriental Medicine, 1672 Yuseongdae-ro, Yuseong-gu, Daejeon, 34054, South Korea
| |
Collapse
|
21
|
Koch M, Ferrarese L, Ben-Yehuda Greenwald M, Werner S. Dose-dependent effects of Nrf2 on the epidermis in chronic skin inflammation. Dis Model Mech 2025; 18:dmm052126. [PMID: 39744884 PMCID: PMC11708820 DOI: 10.1242/dmm.052126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 11/27/2024] [Indexed: 01/11/2025] Open
Abstract
Atopic dermatitis (AD) is a chronic inflammatory skin disease, characterized by an impaired epidermal barrier and immunological alterations. The activity of the cytoprotective NRF2 transcription factor is reduced in the epidermis of AD patients. To determine the functional relevance of this deficiency, we used mice lacking fibroblast growth factor receptors 1 and 2 in keratinocytes (K5-R1/R2 mice), which exhibit several AD-like symptoms. Proteomics analysis of their epidermis revealed reduced Nrf2 activity. This was accompanied by an increase in DNA damage and in the number of senescent cells. Genetic deletion of Nrf2 in keratinocytes of these mice further promoted DNA damage and senescence, but time-limited pharmacological activation of Nrf2 in the skin had a mild protective effect. Surprisingly, long-term genetic activation of Nrf2 in keratinocytes of K5-R1/R2 mice caused strong hyperkeratosis, keratinocyte hyperproliferation, epidermal thickening, increased keratinocyte apoptosis and DNA damage, and altered immune cell composition. These results reveal a complex role of Nrf2 in the epidermis and show the necessity to optimize the duration and intensity of NRF2 activation for the treatment of epidermal alterations in patients with AD.
Collapse
Affiliation(s)
- Michael Koch
- Institute of Molecular Health Sciences, Department of Biology, ETH Zürich, 8093 Zürich, Switzerland
| | - Luca Ferrarese
- Institute of Molecular Health Sciences, Department of Biology, ETH Zürich, 8093 Zürich, Switzerland
| | | | - Sabine Werner
- Institute of Molecular Health Sciences, Department of Biology, ETH Zürich, 8093 Zürich, Switzerland
| |
Collapse
|
22
|
Grieco T, Paolino G, Moliterni E, Chello C, Sernicola A, Egan CG, Morelli M, Nannipieri F, Battaglia S, Accoto M, Tirotta E, Trasciatti S, Bonaretti S, Pellacani G, Calvieri S. Differential Expression of Proteins Involved in Skin Barrier Maintenance and Vitamin D Metabolism in Atopic Dermatitis: A Cross-Sectional, Exploratory Study. Int J Mol Sci 2024; 26:211. [PMID: 39796069 PMCID: PMC11719518 DOI: 10.3390/ijms26010211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 12/18/2024] [Accepted: 12/24/2024] [Indexed: 01/30/2025] Open
Abstract
Atopic dermatitis (AD) is a chronic inflammatory skin disorder influenced by proteins involved in skin barrier maintenance and vitamin D metabolism. Using an intra-patient design, this study compared protein expression in intra-lesional (IL) and peri-lesional (PL) skin biopsies from AD patients and examined associations between protein levels, vitamin D status, and clinical features. Forty-four biopsies from twenty-two AD patients were analyzed using antibody microarrays targeting twelve proteins. IL samples had significantly higher total protein levels than PL samples, with a mean difference of 77.7% (p < 0.001). Several proteins, including cathelicidin, cingulin, occludin, filaggrin, and the vitamin D receptor, were upregulated in IL samples. Patients with vitamin D levels below 30 ng/mL showed higher expression of CYP24A (p = 0.054), alpha-catenin (p = 0.043), and haptoglobin (p = 0.033). Increased EASI scores (≥16) were associated with elevated expression of CYP24A (p = 0.024), CYP27B (p = 0.044), filaggrin (p = 0.027), occludin (p = 0.049), and claudin-1 (p = 0.052). Multivariate regression analysis identified significant correlations between protein expression, skin prick test positivity, and low vitamin D levels. These findings suggest that proteins related to epithelial barrier function and vitamin D metabolism are highly upregulated in IL skin regions, offering potential therapeutic targets for improving both skin barrier function and overall disease severity in AD patients.
Collapse
Affiliation(s)
- Teresa Grieco
- Dermatology Clinic, Department of Clinical Internal, Anesthesiological and Cardiovascular Sciences, Sapienza University of Rome, 00185 Rome, Italy; (T.G.); (E.M.); (C.C.); (A.S.); (G.P.); (S.C.)
| | - Giovanni Paolino
- Unit of Dermatology and Cosmetology, IRCCS University Vita-Salute San Raffaele, 20132 Milan, Italy
| | - Elisa Moliterni
- Dermatology Clinic, Department of Clinical Internal, Anesthesiological and Cardiovascular Sciences, Sapienza University of Rome, 00185 Rome, Italy; (T.G.); (E.M.); (C.C.); (A.S.); (G.P.); (S.C.)
| | - Camilla Chello
- Dermatology Clinic, Department of Clinical Internal, Anesthesiological and Cardiovascular Sciences, Sapienza University of Rome, 00185 Rome, Italy; (T.G.); (E.M.); (C.C.); (A.S.); (G.P.); (S.C.)
| | - Alvise Sernicola
- Dermatology Clinic, Department of Clinical Internal, Anesthesiological and Cardiovascular Sciences, Sapienza University of Rome, 00185 Rome, Italy; (T.G.); (E.M.); (C.C.); (A.S.); (G.P.); (S.C.)
| | | | | | - Fabrizio Nannipieri
- Clinical Research, Abiogen Pharma SpA, 56121 Pisa, Italy; (F.N.); (S.B.); (M.A.); (E.T.)
| | - Santina Battaglia
- Clinical Research, Abiogen Pharma SpA, 56121 Pisa, Italy; (F.N.); (S.B.); (M.A.); (E.T.)
| | - Marina Accoto
- Clinical Research, Abiogen Pharma SpA, 56121 Pisa, Italy; (F.N.); (S.B.); (M.A.); (E.T.)
| | - Erika Tirotta
- Clinical Research, Abiogen Pharma SpA, 56121 Pisa, Italy; (F.N.); (S.B.); (M.A.); (E.T.)
| | | | | | - Giovanni Pellacani
- Dermatology Clinic, Department of Clinical Internal, Anesthesiological and Cardiovascular Sciences, Sapienza University of Rome, 00185 Rome, Italy; (T.G.); (E.M.); (C.C.); (A.S.); (G.P.); (S.C.)
| | - Stefano Calvieri
- Dermatology Clinic, Department of Clinical Internal, Anesthesiological and Cardiovascular Sciences, Sapienza University of Rome, 00185 Rome, Italy; (T.G.); (E.M.); (C.C.); (A.S.); (G.P.); (S.C.)
| |
Collapse
|
23
|
Díez-Madueño K, de la Cueva Dobao P, Torres-Rojas I, Fernández-Gosende M, Hidalgo-Cantabrana C, Coto-Segura P. Gut Dysbiosis and Adult Atopic Dermatitis: A Systematic Review. J Clin Med 2024; 14:19. [PMID: 39797102 PMCID: PMC11721037 DOI: 10.3390/jcm14010019] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 12/16/2024] [Accepted: 12/21/2024] [Indexed: 01/13/2025] Open
Abstract
Background/Objectives: Research on the relationship between gut microbiota (GM) and atopic dermatitis (AD) has seen a growing interest in recent years. The aim of this systematic review was to determine whether differences exist between the GM of adults with AD and that of healthy adults (gut dysbiosis). Methods: We conducted a systematic review based on the PRISMA guidelines (Preferred Reporting Items for Systematic Reviews and Meta-Analyses). The search was performed using PubMed, EMBASE, and Web of Science. Observational and interventional studies were analyzed. Results: Although the studies showed heterogeneous results, some distinguishing characteristics were found in the intestinal microbial composition of adults with dermatitis. Even though no significant differences in diversity were found between healthy and affected adults, certain microorganisms, such as Bacteroidales, Enterobacteriaceae, and Clostridium (perfringens), were more characteristic of the fecal microbiota in adults with AD. Healthy individuals exhibited lower abundances of aerobic bacteria and higher abundances of short-chain fatty acid-producing species and polyamines. Clinical trials showed that the consumption of probiotics (Bifidobacterium and/or Lactobacillus), fecal microbiota transplants, and balneotherapy modified the fecal microbiota composition of participants and were associated with significant improvements in disease management. Conclusions: In anticipation of forthcoming clinical trials, it is essential to conduct meta-analyses that comprehensively evaluate the effectiveness and safety of interventions designed to modify intestinal flora in the context of AD. Preliminary evidence suggests that certain interventions may enhance adult AD management.
Collapse
Affiliation(s)
- Kevin Díez-Madueño
- Dermatology Department, Hospital Universitario Infanta Leonor, Complutense University of Madrid, 28040 Madrid, Spain;
- School of Medicine, Complutense University of Madrid, 28040 Madrid, Spain
| | - Pablo de la Cueva Dobao
- Dermatology Department, Hospital Universitario Infanta Leonor, Complutense University of Madrid, 28040 Madrid, Spain;
- School of Medicine, Complutense University of Madrid, 28040 Madrid, Spain
| | - Isabel Torres-Rojas
- Allergy Department, Hospital Universitario Infanta Sofía, 28702 Alcobendas, Spain;
| | | | | | - Pablo Coto-Segura
- Dermatology Department, Hospital Vital Álvarez Buylla, 33611 Mieres, Spain;
| |
Collapse
|
24
|
Houtsaeger C, Pasmans F, Claes I, Vandenabeele S, Haesebrouck F, Lebeer S, Boyen F. The role of the microbiome in allergic dermatitis-related otitis externa: a multi-species comparative review. Front Vet Sci 2024; 11:1413684. [PMID: 39736936 PMCID: PMC11683847 DOI: 10.3389/fvets.2024.1413684] [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: 04/07/2024] [Accepted: 11/27/2024] [Indexed: 01/01/2025] Open
Abstract
The external ear canal, characterized by species-specific structural and physiological differences, maintains a hostile environment that prevents microbial overgrowth and foreign body entry, supported by factors such as temperature, pH, humidity, and cerumen with antimicrobial properties. This review combines several studies on the healthy ear canal's structure and physiology with a critical approach to the potential existence of an ear microbiome. We use a comparative multi-species approach to explore how allergic conditions alter the ear canal microenvironment and cerumen in different mammalian species, promoting pathogen colonization. We propose a pathogenetic model in which allergic conditions disrupt the antimicrobial environment of the EEC, creating circumstances favorable for facultative pathogenic micro-organisms like Staphylococcus and Malassezia species, leading to otitis externa (OE). A better understanding of the underpinning mechanisms may lead to innovative approaches to disease mitigation.
Collapse
Affiliation(s)
- Cyrelle Houtsaeger
- Department of Pathobiology Pharmacology and Zoological Medicine, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
- YUN NV, Niel, Belgium
| | - Frank Pasmans
- Department of Pathobiology Pharmacology and Zoological Medicine, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Ingmar Claes
- YUN NV, Niel, Belgium
- Department of Bioscience Engineering, University of Antwerp, Antwerp, Belgium
| | - Sophie Vandenabeele
- Department of Small Animal Medicine and Clinical Biology, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Freddy Haesebrouck
- Department of Pathobiology Pharmacology and Zoological Medicine, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Sarah Lebeer
- Department of Bioscience Engineering, University of Antwerp, Antwerp, Belgium
| | - Filip Boyen
- Department of Pathobiology Pharmacology and Zoological Medicine, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| |
Collapse
|
25
|
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
|
26
|
Sun N, Ogulur I, Mitamura Y, Yazici D, Pat Y, Bu X, Li M, Zhu X, Babayev H, Ardicli S, Ardicli O, D'Avino P, Kiykim A, Sokolowska M, van de Veen W, Weidmann L, Akdis D, Ozdemir BG, Brüggen MC, Biedermann L, Straumann A, Kreienbühl A, Guttman-Yassky E, Santos AF, Del Giacco S, Traidl-Hoffmann C, Jackson DJ, Wang DY, Lauerma A, Breiteneder H, Zhang L, O'Mahony L, Pfaar O, O'Hehir R, Eiwegger T, Fokkens WJ, Cabanillas B, Ozdemir C, Kistler W, Bayik M, Nadeau KC, Torres MJ, Akdis M, Jutel M, Agache I, Akdis CA. The epithelial barrier theory and its associated diseases. Allergy 2024; 79:3192-3237. [PMID: 39370939 DOI: 10.1111/all.16318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 08/28/2024] [Accepted: 09/03/2024] [Indexed: 10/08/2024]
Abstract
The prevalence of many chronic noncommunicable diseases has been steadily rising over the past six decades. During this time, over 350,000 new chemical substances have been introduced to the lives of humans. In recent years, the epithelial barrier theory came to light explaining the growing prevalence and exacerbations of these diseases worldwide. It attributes their onset to a functionally impaired epithelial barrier triggered by the toxicity of the exposed substances, associated with microbial dysbiosis, immune system activation, and inflammation. Diseases encompassed by the epithelial barrier theory share common features such as an increased prevalence after the 1960s or 2000s that cannot (solely) be accounted for by the emergence of improved diagnostic methods. Other common traits include epithelial barrier defects, microbial dysbiosis with loss of commensals and colonization of opportunistic pathogens, and circulating inflammatory cells and cytokines. In addition, practically unrelated diseases that fulfill these criteria have started to emerge as multimorbidities during the last decades. Here, we provide a comprehensive overview of diseases encompassed by the epithelial barrier theory and discuss evidence and similarities for their epidemiology, genetic susceptibility, epithelial barrier dysfunction, microbial dysbiosis, and tissue inflammation.
Collapse
Affiliation(s)
- Na Sun
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
- SKL of Marine Food Processing & Safety Control, National Engineering Research Center of Seafood, School of Food Science and Technology, Dalian Polytechnic University, Dalian, P. R. China
| | - Ismail Ogulur
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Yasutaka Mitamura
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Duygu Yazici
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Yagiz Pat
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Xiangting Bu
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Manru Li
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Xueyi Zhu
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Huseyn Babayev
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Sena Ardicli
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
- Department of Genetics, Faculty of Veterinary Medicine, Bursa Uludag University, Bursa, Turkey
| | - Ozge Ardicli
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
- Division of Food Processing, Milk and Dairy Products Technology Program, Karacabey Vocational School, Bursa Uludag University, Bursa, Turkey
| | - Paolo D'Avino
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Ayca Kiykim
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
- Department of Pediatrics, Division of Pediatric Allergy and Immunology, Cerrahpasa School of Medicine, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Milena Sokolowska
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Willem van de Veen
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Lukas Weidmann
- Department of Nephrology, University Hospital Zurich, Zurich, Switzerland
| | - Deniz Akdis
- Department of Cardiology, University Hospital Zurich, Zurich, Switzerland
| | | | - Marie Charlotte Brüggen
- Christine Kühne-Center for Allergy Research and Education (CK-CARE), Davos, Switzerland
- Faculty of Medicine, University of Zurich, Zurich, Switzerland
- Department of Dermatology, University Hospital Zurich, Zurich, Switzerland
| | - Luc Biedermann
- Department of Gastroenterology and Hepatology, University Hospital Zurich, Zurich, Switzerland
| | - Alex Straumann
- Department of Gastroenterology and Hepatology, University Hospital Zurich, Zurich, Switzerland
| | - Andrea Kreienbühl
- Department of Gastroenterology and Hepatology, University Hospital Zurich, Zurich, Switzerland
| | - Emma Guttman-Yassky
- Department of Dermatology, and Laboratory of Inflammatory Skin Diseases, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Alexandra F Santos
- Department of Women and Children's Health (Pediatric Allergy), School of Life Course Sciences, Faculty of Life Sciences and Medicine, King's College London, London, UK
- Children's Allergy Service, Evelina London Children's Hospital, Guy's and St. Thomas' Hospital, London, UK
- Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, King's College London, London, UK
| | - Stefano Del Giacco
- Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, Italy
| | | | - David J Jackson
- Guy's Severe Asthma Centre, Guy's Hospital, Guy's & St Thomas' NHS Trust, London, UK
- School of Immunology & Microbial Sciences, King's College London, London, UK
| | - De-Yun Wang
- Department of Otolaryngology, Infectious Diseases Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, National University Health System, Singapore City, Singapore
| | - Antti Lauerma
- Department of Dermatology, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Heimo Breiteneder
- Department of Pathophysiology and Allergy Research, Medical University of Vienna, Vienna, Austria
| | - Luo Zhang
- Department of Otolaryngology Head and Neck Surgery, Beijing Tongren Hospital, Capital Medical University, Beijing, China
- Beijing Laboratory of Allergic Diseases and Beijing Key Laboratory of Nasal Diseases, Beijing Institute of Otolaryngology, Beijing, China
| | - Liam O'Mahony
- Department of Medicine and School of Microbiology, University College Cork, Cork, Ireland
- APC Microbiome Ireland, Cork, Ireland
| | - Oliver Pfaar
- Department of Otorhinolaryngology, Head and Neck Surgery, Section of Rhinology and Allergy, University Hospital Marburg, Philipps-Universität Marburg, Marburg, Germany
| | - Robyn O'Hehir
- Allergy, Asthma & Clinical Immunology, The Alfred Hospital, Melbourne, Victoria, Australia
- Department of Immunology, School of Translational Medicine, Monash University, Melbourne, Victoria, Australia
| | - Thomas Eiwegger
- Translational Medicine Program, Research Institute, Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Immunology, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
- Karl Landsteiner University of Health Sciences, Krems an der Donau, Austria
- Department of Pediatric and Adolescent Medicine, University Hospital St. Pölten, St. Pölten, Austria
| | - Wytske J Fokkens
- Department of Otorhinolaryngology & Head and Neck Surgery, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Beatriz Cabanillas
- Department of Allergy, Instituto de Investigación Biosanitaria Hospital 12 de Octubre (imas12), Madrid, Spain
| | - Cevdet Ozdemir
- Department of Pediatric Basic Sciences, Institute of Child Health, Istanbul University, Istanbul, Turkey
- Istanbul Faculty of Medicine, Department of Pediatrics, Division of Pediatric Allergy and Immunology, Istanbul University, Istanbul, Turkey
| | - Walter Kistler
- Department of Sports Medicine, Davos Hospital, Davos, Switzerland
- Swiss Research Institute for Sports Medicine (SRISM), Davos, Switzerland
- Medical Committee International Ice Hockey Federation (IIHF), Zurich, Switzerland
| | - Mahmut Bayik
- Department of Internal Medicine and Hematology, Marmara University, Istanbul, Turkey
| | - Kari C Nadeau
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Maria J Torres
- Allergy Unit, IBIMA-Hospital Regional Universitario de Málaga-ARADyAL, UMA, Málaga, Spain
| | - Mübeccel Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Marek Jutel
- Department of Clinical Immunology, Wrocław Medical University, Wroclaw, Poland
| | - Ioana Agache
- Faculty of Medicine, Department of Allergy and Clinical Immunology, Transylvania University, Brasov, Romania
| | - Cezmi A Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| |
Collapse
|
27
|
Dajnoki Z, Kapitány A, Eyerich K, Eyerich S, Törőcsik D, Szegedi A. Topographical variations in the skin barrier and their role in disease pathogenesis. J Eur Acad Dermatol Venereol 2024. [PMID: 39607016 DOI: 10.1111/jdv.20463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Accepted: 10/09/2024] [Indexed: 11/29/2024]
Abstract
The skin barrier can be divided into at least four functional units: chemical, microbial, physical and immunological barriers. The chemical and microbial barriers have previously been shown to exhibit different characteristics in topographically distinct skin regions. There is increasing evidence that the physical and immunological barriers also show marked variability in different areas of the skin. Here, we review recent data on the topographical variations of skin barrier components, the contribution of these variations to the homeostatic function of the skin and their impact on the pathogenesis of specific immune-mediated skin diseases (such as atopic dermatitis and papulopustular rosacea). Recognition of these topographical barrier differences will improve our understanding of skin homeostasis and disease pathogenesis and provide a basis for body site-specific targeted therapies.
Collapse
Affiliation(s)
- Z Dajnoki
- Department of Dermatology, MTA Centre of Excellence, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
- HUN-REN-UD Allergology Research Group, Debrecen, Hungary
| | - A Kapitány
- Department of Dermatology, MTA Centre of Excellence, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
- HUN-REN-UD Allergology Research Group, Debrecen, Hungary
| | - K Eyerich
- Department of Dermatology and Venerology, Medical Center, University of Freiburg, Freiburg, Germany
| | - S Eyerich
- Center for Allergy and Environment (ZAUM), Technical University and Helmholtz Center Munich, Munich, Germany
| | - D Törőcsik
- Department of Dermatology, MTA Centre of Excellence, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
- HUN-REN-UD Allergology Research Group, Debrecen, Hungary
| | - A Szegedi
- Department of Dermatology, MTA Centre of Excellence, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
- HUN-REN-UD Allergology Research Group, Debrecen, Hungary
| |
Collapse
|
28
|
Kistler W, Villiger M, Villiger B, Yazici D, Pat Y, Mitamura Y, Ardicli S, Skolnick S, Dhir R, Akdis M, Nadeau K, Ogulur I, Akdis CA. Epithelial barrier theory in the context of nutrition and environmental exposure in athletes. Allergy 2024; 79:2912-2923. [PMID: 39011970 DOI: 10.1111/all.16221] [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: 03/27/2024] [Revised: 06/18/2024] [Accepted: 06/28/2024] [Indexed: 07/17/2024]
Abstract
Exposure to toxic substances, introduced into our daily lives during industrialization and modernization, can disrupt the epithelial barriers in the skin, respiratory, and gastrointestinal systems, leading to microbial dysbiosis and inflammation. Athletes and physically active individuals are at increased risk of exposure to agents that damage the epithelial barriers and microbiome, and their extreme physical exercise exerts stress on many organs, resulting in tissue damage and inflammation. Epithelial barrier-damaging substances include surfactants and enzymes in cleaning products, laundry and dishwasher detergents, chlorine in swimming pools, microplastics, air pollutants such as ozone, particulate matter, and diesel exhaust. Athletes' high-calorie diet often relies on processed foods that may contain food emulsifiers and other additives that may cause epithelial barrier dysfunction and microbial dysbiosis. The type of the material used in the sport equipment and clothing and their extensive exposure may increase the inflammatory effects. Excessive travel-related stress, sleep disturbances and different food and microbe exposure may represent additional factors. Here, we review the detrimental impact of toxic agents on epithelial barriers and microbiome; bring a new perspective on the factors affecting the health and performance of athletes and physically active individuals.
Collapse
Affiliation(s)
- Walter Kistler
- Medical Committee International Ice Hockey Federation, Zürich, Switzerland
- Swiss Research Institute for Sports Medicine (SRISM), Davos, Switzerland
- Department of Sports Medicine, Davos Hospital, Davos, Switzerland
| | - Michael Villiger
- Swiss Research Institute for Sports Medicine (SRISM), Davos, Switzerland
- Department of Sports Medicine, Davos Hospital, Davos, Switzerland
| | - Beat Villiger
- Swiss Research Institute for Sports Medicine (SRISM), Davos, Switzerland
- Department of Sports Medicine, Davos Hospital, Davos, Switzerland
| | - Duygu Yazici
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Yagiz Pat
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Yasutaka Mitamura
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Sena Ardicli
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Stephen Skolnick
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
- Seed Health Inc., Los Angeles, California, USA
| | - Raja Dhir
- Seed Health Inc., Los Angeles, California, USA
| | - Mübeccel Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Kari Nadeau
- Department of Environmental Health, T.H. Chan School of Public Health, Harvard University, Boston, Massachusetts, USA
| | - Ismail Ogulur
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Cezmi A Akdis
- Swiss Research Institute for Sports Medicine (SRISM), Davos, Switzerland
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| |
Collapse
|
29
|
Buchheit KM, Shaw D, Chupp G, Lehtimaki L, Heffler E, Finney-Hayward T, Zangrilli J, Kwiatek J, Siddiqui S, Roufosse F, Thamboo A, West N, Vichiendilokkul A, Hellings PW, Peters A, Howarth PH. Interleukin-5 as a pleiotropic cytokine orchestrating airway type 2 inflammation: Effects on and beyond eosinophils. Allergy 2024; 79:2662-2679. [PMID: 39359069 DOI: 10.1111/all.16303] [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: 05/07/2024] [Revised: 08/05/2024] [Accepted: 08/20/2024] [Indexed: 10/04/2024]
Abstract
Interleukin (IL)-5 is the key cytokine in the maturation, activation, proliferation, migration and survival of eosinophils, which are key effector cells in many upper and lower airway diseases. Through its effects on eosinophils, IL-5 indirectly contributes to various pathophysiological processes including tissue damage, repair and remodelling. Understanding the importance of IL-5 in eosinophil-associated diseases led to the development of anti-IL-5 therapies, which provide clinical benefits across a range of conditions. However, recent evidence suggests that eosinophil-depletion alone may not account for all of the therapeutic effects of anti-IL-5 therapy and that IL-5 may also contribute to disease independently of its effects on eosinophils. Indeed, evidence from ex vivo studies and targeted therapy in vivo demonstrates that IL-5 and its inhibition affects a much broader range of cells beyond eosinophils, including epithelial cells, plasma cells, mast cells, basophils, neutrophils, type 2 innate lymphoid cells, T regulatory cells and fibroblasts. This review will provide an update on the evidence supporting the breadth of IL-5 biology relevant to disease pathogenesis beyond eosinophil-associated inflammation, where there is a need for additional insight, and the clinical implications of a more central role of IL-5 in type 2 inflammation.
Collapse
Affiliation(s)
- Kathleen M Buchheit
- Division of Allergy and Clinical Immunology, AERD Center, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Dominick Shaw
- Department of Respiratory Medicine and Thoracic Surgery, Institute for Lung Health, Glenfield Hospital, Leicester, UK
| | - Geoffrey Chupp
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, Connecticut, USA
| | - Lauri Lehtimaki
- Faculty of Medicine and Health Technology, Allergy Centre, Tampere University Hospital, Tampere University, Tampere, Finland
| | - Enrico Heffler
- Department of Biomedical Sciences, Humanitas University, Milan, Italy
- Personalized Medicine, Asthma and Allergy Clinic, IRCCS Humanitas Research Hospital, Milan, Italy
| | | | | | - Justin Kwiatek
- US Medical Affairs, GSK, Collegeville, Pennsylvania, USA
| | - Salman Siddiqui
- National Heart and Lung Institute, NIHR Imperial Biomedical Research Centre, Imperial College London, London, UK
| | - Florence Roufosse
- Department of Internal Medicine, Erasme Hospital, Université Libre de Bruxelles, Brussels, Belgium
| | - Andrew Thamboo
- Division of Otolaryngology-Head and Neck Surgery, Department of Surgery, University of British Columbia, Vancouver, British Columbia, Canada
| | - Nicholas West
- School of Pharmacy and Medical Sciences, Griffith University, Southport, Queensland, Australia
| | | | - Peter W Hellings
- Department of Otorhinolaryngology, Head and Neck Surgery, University Hospitals Leuven, Leuven, Belgium
- Upper Airways Disease Research Unit, University of Ghent, Ghent, Belgium
| | - Anju Peters
- Division of Allergy and Immunology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Peter H Howarth
- Global Medical Affairs, Respiratory Specialty Care, GSK, London, UK
| |
Collapse
|
30
|
Narla S, Silverberg JI. Atopic Dermatitis: A Disorder of both Adults and Children with Varying Longitudinal Course. Dermatol Clin 2024; 42:513-518. [PMID: 39278704 DOI: 10.1016/j.det.2024.05.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/18/2024]
Abstract
The longitudinal course of atopic dermatitis (AD) is heterogeneous and complex. While previously thought to be a childhood disorder, recent studies demonstrated that childhood-onset AD may take several different courses that may involve persistence into adulthood becoming a lifelong condition. Other patients only demonstrated adult-onset AD. Different factors may play a role in the timing of AD onset. Assessing the longitudinal course also involves understanding the changing temporal pattern of AD. Understanding the dynamic course of AD is important in identifying individualized treatment recommendations for patients.
Collapse
Affiliation(s)
- Shanthi Narla
- Department of Dermatology, Medical College of Wisconsin, Office A3698, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA
| | - Jonathan I Silverberg
- Department of Dermatology, The George Washington University School of Medicine and Health Sciences, Suite 2B-430, 2150 Pennsylvania Avenue, Washington, DC 20037, USA.
| |
Collapse
|
31
|
Macura B, Kiecka A, Szczepanik M. Intestinal permeability disturbances: causes, diseases and therapy. Clin Exp Med 2024; 24:232. [PMID: 39340718 PMCID: PMC11438725 DOI: 10.1007/s10238-024-01496-9] [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: 08/20/2024] [Accepted: 09/22/2024] [Indexed: 09/30/2024]
Abstract
Nowadays, a pathological increase in the permeability of the intestinal barrier (the so-called leaky gut) is increasingly being diagnosed. This condition can be caused by various factors, mainly from the external environment. Damage to the intestinal barrier entails a number of adverse phenomena: dysbiosis, translocation of microorganisms deep into the intestinal tissue, immune response, development of chronic inflammation. These phenomena can ultimately lead to a vicious cycle that promotes the development of inflammation and further damage to the barrier. Activated immune cells in mucosal tissues with broken barriers can migrate to other organs and negatively affect their functioning. Damaged intestinal barrier can facilitate the development of local diseases such as irritable bowel disease, inflammatory bowel disease or celiac disease, but also the development of systemic inflammatory diseases such as rheumatoid arthritis, ankylosing spondylitis, hepatitis, and lupus erythematosus, neurodegenerative or psychiatric conditions, or metabolic diseases such as diabetes or obesity. However, it must be emphasized that the causal links between a leaky gut barrier and the onset of certain diseases often remain unclear and require in-depth research. In light of recent research, it becomes crucial to prevent damage to the intestinal barrier, as well as to develop therapies for the barrier when it is damaged. This paper presents the current state of knowledge on the causes, health consequences and attempts to treat excessive permeability of the intestinal barrier.
Collapse
Affiliation(s)
- Barbara Macura
- Faculty of Health Sciences, Institute of Physiotherapy, Chair of Biomedical Sciences, Jagiellonian University Medical College, Kopernika 7a, 31-034, Kraków, Poland.
| | - Aneta Kiecka
- Faculty of Health Sciences, Institute of Physiotherapy, Chair of Biomedical Sciences, Jagiellonian University Medical College, Kopernika 7a, 31-034, Kraków, Poland
| | - Marian Szczepanik
- Faculty of Health Sciences, Institute of Physiotherapy, Chair of Biomedical Sciences, Jagiellonian University Medical College, Kopernika 7a, 31-034, Kraków, Poland
| |
Collapse
|
32
|
Özdemіr E, Öksüz L. Effect of Staphylococcus aureus colonization and immune defects on the pathogenesis of atopic dermatitis. Arch Microbiol 2024; 206:410. [PMID: 39302484 DOI: 10.1007/s00203-024-04134-w] [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: 07/27/2024] [Revised: 08/26/2024] [Accepted: 09/07/2024] [Indexed: 09/22/2024]
Abstract
Atopic dermatitis (AD) is a common and recurrent skin disease characterized by skin barrier dysfunction, inflammation and chronic pruritus, with wide heterogeneity in terms of age of onset, clinical course and persistence over the lifespan. Although the pathogenesis of the disease are unclear, epidermal barrier dysfunction, immune and microbial dysregulation, and environmental factors are known to be critical etiologies in AD pathology. The skin microbiota represents an ecosystem consisting of numerous microbial species that interact with each other as well as host epithelial cells and immune cells. Although the skin microbiota benefits the host by supporting the basic functions of the skin and preventing the colonization of pathogens, disruption of the microbial balance (dysbiosis) can cause skin diseases such as AD. Although AD is a dermatological disease, recent evidence has shown that changes in microbiota composition in the skin and intestine contribute to the pathogenesis of AD. Environmental factors that contribute to skin barrier dysfunction and microbial dysbiosis in AD include allergens, diet, irritants, air pollution, epigenetics and microbial exposure. Knowing the microbial combination of intestin, as well as the genetic and epigenetic determinants associated with the development of autoantibodies, may help elucidate the pathophysiology of the disease. The skin of patients with AD is characterized by microbial dysbiosis as a result of reduced microbial diversity and overgrowth of the pathogens such as Staphylococcus aureus. Recent studies have revealed the importance of building a strong immune response against microorganisms during childhood and new mechanisms of microbial community dynamics in modulating the skin microbiome. Numerous microorganisms are reported to modulate host response through communication with keratinocytes, specific immune cells and adipocytes to improve skin health and barrier function. This growing insight into bioactive substances in the skin microbiota has led to novel biotherapeutic approaches targeting the skin surface for the treatment of AD. This review will provide an updated overview of the skin microbiota in AD and its complex interaction with immune response mechanisms, as well as explore possible underlying mechanisms in the pathogenesis of AD and provide insights into new therapeutic developments for the treatment of AD. It also focuses on restoring skin microbial homeostasis, aiming to reduce inflammation by repairing the skin barrier.
Collapse
Affiliation(s)
- Evrim Özdemіr
- Department of Medical Microbiology, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey.
- Institute of Graduate Studies in Health Sciences, Istanbul University, Istanbul, Turkey.
| | - Lütfiye Öksüz
- Department of Medical Microbiology, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| |
Collapse
|
33
|
Sampson HA. The riddle of response to cutaneous allergen exposure in patients with atopic dermatitis. Ann Allergy Asthma Immunol 2024; 133:244-251. [PMID: 38740132 DOI: 10.1016/j.anai.2024.05.005] [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: 04/04/2024] [Revised: 04/29/2024] [Accepted: 05/02/2024] [Indexed: 05/16/2024]
Abstract
The skin is the largest immunologic organ in the body and contains immune cells that play a role in both food allergen sensitization and desensitization. The dual allergen exposure hypothesis posits that sensitization to food allergens may occur with cutaneous exposure on inflamed skin, eg, atopic dermatitis, but early oral consumption generally leads to tolerance. However, only one-third of children with atopic dermatitis develop a food allergy, suggesting that there is a more complex mechanism for allergen sensitization. Emerging evidence suggests that the outcome of cutaneous allergen exposure is context-dependent and largely influenced by the state of the skin barrier with healthy skin promoting natural tolerance. Current research supports the ability to induce desensitization through repeated application of allergens to the skin, known as epicutaneous immunotherapy. Preclinical research with an occlusive patch has demonstrated a significantly reduced T-helper cell type 2-driven immunologic response when applied to intact, uninflamed skin and induction of a unique population of regulatory T cells that express a broader range of homing receptors, which may be able to maintain sustained protection. In clinical studies of children aged 1 through 11 years with a peanut allergy, epicutaneous immunotherapy with an occlusive patch led to significant desensitization with no major differences in efficacy or safety between children with and without atopic dermatitis. These data begin to answer the conundrum of how allergens that are applied to the skin can lead to both sensitization and desensitization, and future studies should enable us to optimize the power of the skin as a complex immunologic organ to treat allergic, autoimmune, and autoinflammatory disorders.
Collapse
Affiliation(s)
- Hugh A Sampson
- Division of Allergy and Immunology, Department of Pediatrics, Jaffe Food Allergy Research Institute, Icahn School of Medicine at Mount Sinai, New York, New York.
| |
Collapse
|
34
|
Wang R, Shu RR, Seldin L. Noncanonical functions of adhesion proteins in inflammation. Am J Physiol Cell Physiol 2024; 327:C505-C515. [PMID: 38981610 PMCID: PMC11427013 DOI: 10.1152/ajpcell.00292.2024] [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: 05/02/2024] [Revised: 06/24/2024] [Accepted: 07/01/2024] [Indexed: 07/11/2024]
Abstract
Cell adhesion proteins localize to epithelial and endothelial cell membranes to form junctional complexes between neighboring cells or between cells and the underlying basement membrane. The structural and functional integrities of these junctions are critical to establish cell polarity and maintain tissue barrier function, while also facilitating leukocyte migration and adhesion to sites of inflammation. In addition to their adhesive properties, however, junctional proteins can also serve important noncanonical functions in inflammatory signaling and transcriptional regulation. Intriguingly, recent work has unveiled novel roles for cell adhesion proteins as both signaling initiators and downstream targets during inflammation. In this review, we discuss both the traditional functions of junction proteins in cell adhesion and tissue barrier function as well as their noncanonical signaling roles that have been implicated in facilitating diverse inflammatory pathologies.
Collapse
Affiliation(s)
- Ruochong Wang
- Department of Cell Biology, Emory University School of Medicine, Atlanta, Georgia, United States
| | - Raphael R Shu
- Department of Cell Biology, Emory University School of Medicine, Atlanta, Georgia, United States
| | - Lindsey Seldin
- Department of Cell Biology, Emory University School of Medicine, Atlanta, Georgia, United States
- Department of Dermatology, Emory University School of Medicine, Atlanta, Georgia, United States
- Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia, United States
- Atlanta Veterans Affairs Medical Center, Decatur, Georgia, United States
| |
Collapse
|
35
|
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
|
36
|
Marsch P, Rajagopal N, Nangia S. Biophysics of claudin proteins in tight junction architecture: Three decades of progress. Biophys J 2024; 123:2363-2378. [PMID: 38859584 PMCID: PMC11365114 DOI: 10.1016/j.bpj.2024.06.010] [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: 01/17/2024] [Revised: 05/19/2024] [Accepted: 06/07/2024] [Indexed: 06/12/2024] Open
Abstract
Tight junctions are cell-cell adhesion complexes that act as gatekeepers of the paracellular space. Formed by several transmembrane proteins, the claudin family performs the primary gate-keeping function. The claudin proteins form charge and size-selective diffusion barriers to maintain homeostasis across endothelial and epithelial tissue. Of the 27 known claudins in mammals, some are known to seal the paracellular space, while others provide selective permeability. The differences in permeability arise due to the varying expression levels of claudins in each tissue. The tight junctions are observed as strands in freeze-fracture electron monographs; however, at the molecular level, tight junction strands form when multiple claudin proteins assemble laterally (cis assembly) within a cell and head-on (trans assembly) with claudins of the adjacent cell in a zipper-like architecture, closing the gap between the neighboring cells. The disruption of tight junctions caused by changing claudin expression levels or mutations can lead to diseases. Therefore, knowledge of the molecular architecture of the tight junctions and how that is tied to tissue-specific function is critical for fighting diseases. Here, we review the current understanding of the tight junctions accrued over the last three decades from experimental and computational biophysics perspectives.
Collapse
Affiliation(s)
- Patrick Marsch
- Department of Biomedical and Chemical Engineering, Syracuse University, Syracuse, New York
| | - Nandhini Rajagopal
- Department of Biomedical and Chemical Engineering, Syracuse University, Syracuse, New York
| | - Shikha Nangia
- Department of Biomedical and Chemical Engineering, Syracuse University, Syracuse, New York.
| |
Collapse
|
37
|
Li L, Liu Y, Chang R, Ye T, Li Z, Huang R, Wang Z, Deng J, Xia H, Yang Y, Huang Y. Dermal Injection of Recombinant Filaggrin-2 Ameliorates UVB-Induced Epidermal Barrier Dysfunction and Photoaging. Antioxidants (Basel) 2024; 13:1002. [PMID: 39199247 PMCID: PMC11351670 DOI: 10.3390/antiox13081002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 08/15/2024] [Accepted: 08/17/2024] [Indexed: 09/01/2024] Open
Abstract
The epidermal barrier is vital for protecting the skin from environmental stressors and ultraviolet (UV) radiation. Filaggrin-2 (FLG2), a critical protein in the stratum corneum, plays a significant role in maintaining skin barrier homeostasis. However, the precise role of FLG2 in mitigating the adverse effects of UV-induced barrier disruption and photoaging remains poorly understood. In this study, we revealed that UVB exposure resulted in a decreased expression of FLG2 in HaCaT keratinocytes, which correlated with a compromised barrier function. The administration of recombinant filaggrin-2 (rFLG2) enhanced keratinocyte differentiation, bolstered barrier integrity, and offered protection against apoptosis and oxidative stress induced by UVB irradiation. Furthermore, in a UV-induced photodamage murine model, the dermal injection of rFLG2 facilitated the enhanced restoration of the epidermal barrier, decreased oxidative stress and inflammation, and mitigated the collagen degradation that is typical of photoaging. Collectively, our findings suggested that targeting FLG2 could be a strategic approach to prevent and treat skin barrier dysfunction and combat the aging effects associated with photoaging. rFLG2 emerges as a potentially viable therapy for maintaining skin health and preventing skin aging processes amplified by photodamage.
Collapse
Affiliation(s)
- Lu Li
- Department of Cell Biology, Jinan University, Guangzhou 510632, China; (L.L.); (Y.L.); (R.C.); (T.Y.); (R.H.); (Z.W.); (J.D.)
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou 510632, China
| | - Yuan Liu
- Department of Cell Biology, Jinan University, Guangzhou 510632, China; (L.L.); (Y.L.); (R.C.); (T.Y.); (R.H.); (Z.W.); (J.D.)
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou 510632, China
| | - Ruxue Chang
- Department of Cell Biology, Jinan University, Guangzhou 510632, China; (L.L.); (Y.L.); (R.C.); (T.Y.); (R.H.); (Z.W.); (J.D.)
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou 510632, China
| | - Tao Ye
- Department of Cell Biology, Jinan University, Guangzhou 510632, China; (L.L.); (Y.L.); (R.C.); (T.Y.); (R.H.); (Z.W.); (J.D.)
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou 510632, China
| | - Ziyi Li
- TYRAN Cosmetics Innovation Research Institute, Jinan University, Guangzhou 511447, China; (Z.L.); (H.X.)
| | - Rufei Huang
- Department of Cell Biology, Jinan University, Guangzhou 510632, China; (L.L.); (Y.L.); (R.C.); (T.Y.); (R.H.); (Z.W.); (J.D.)
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou 510632, China
| | - Zhaoyang Wang
- Department of Cell Biology, Jinan University, Guangzhou 510632, China; (L.L.); (Y.L.); (R.C.); (T.Y.); (R.H.); (Z.W.); (J.D.)
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou 510632, China
| | - Jingxian Deng
- Department of Cell Biology, Jinan University, Guangzhou 510632, China; (L.L.); (Y.L.); (R.C.); (T.Y.); (R.H.); (Z.W.); (J.D.)
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou 510632, China
| | - Huan Xia
- TYRAN Cosmetics Innovation Research Institute, Jinan University, Guangzhou 511447, China; (Z.L.); (H.X.)
| | - Yan Yang
- Department of Cell Biology, Jinan University, Guangzhou 510632, China; (L.L.); (Y.L.); (R.C.); (T.Y.); (R.H.); (Z.W.); (J.D.)
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou 510632, China
- National Engineering Research Center of Genetic Medicine, Guangzhou 510632, China
- Guangdong Province Key Laboratory of Bioengineering Medicine, Guangzhou 510632, China
| | - Yadong Huang
- Department of Cell Biology, Jinan University, Guangzhou 510632, China; (L.L.); (Y.L.); (R.C.); (T.Y.); (R.H.); (Z.W.); (J.D.)
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Jinan University, Guangzhou 510632, China
- National Engineering Research Center of Genetic Medicine, Guangzhou 510632, China
- Guangdong Province Key Laboratory of Bioengineering Medicine, Guangzhou 510632, China
| |
Collapse
|
38
|
Berekmeri A, Macleod T, Hyde I, Ojak GJ, Mann C, Kramer D, Stacey M, Wittmann M. Epidermal proteomics demonstrates Elafin as a psoriasis-specific biomarker and highlights increased anti-inflammatory activity around psoriatic plaques. J Eur Acad Dermatol Venereol 2024. [PMID: 39157924 DOI: 10.1111/jdv.20289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Accepted: 07/08/2024] [Indexed: 08/20/2024]
Abstract
BACKGROUND Eczema and psoriasis are common diseases. Despite both showing active epidermal contribution to the inflammatory process, their molecular aetiology and pathological mechanisms are different. OBJECTIVE Further molecular insight into these differences is therefore needed to enable effective future diagnostic and treatment strategies. The majority of our mechanistic and clinical understanding of psoriasis and eczema is derived from RNA, immunohistology and whole skin biopsy data. METHODS In this study, non-invasive epidermal sampling of lesional, perilesional and non-lesional skin from diseased and healthy skin was used to perform an in depth proteomic analysis of epidermal proteins. RESULTS Our findings confirmed the psoriasis-associated cytokine IL-36γ as an excellent protein biomarker for lesional psoriasis. However, ELISA and ROC curve analysis of 53 psoriasis and 42 eczema derived samples showed that the sensitivity and specificity were outperformed by elastase-specific protease inhibitor, elafin. Of note, elafin was also found upregulated in non-lesional psoriatic skin at non-predilection sites demonstrating inherent differences between the non-involved skin of healthy and psoriatic individuals. Mass spectrometry and ELISA analysis also demonstrated the upregulation of the anti-inflammatory molecule IL-37 in psoriatic perilesional but not lesional skin. The high expression of IL-37 surrounding psoriatic plaque may contribute to the sharp demarcation of inflammatory morphology changes observed in psoriasis. This finding was also specific for psoriasis and not seen in atopic dermatitis or autoimmune blistering perilesional skin. Our results confirm IL-36γ and add elafin as robust, hallmark molecules distinguishing psoriasis and eczema-associated inflammation even in patients under systemic treatment. CONCLUSIONS Overall, these findings highlight the potential of epidermal non-invasive sampling and proteomic analysis to increase our diagnostic and pathophysiologic understanding of skin diseases. Moreover, the identification of molecular differences in healthy-looking skin between patients and healthy controls highlights potential disease susceptibility markers and proteins involved in the initial stages of disease.
Collapse
Affiliation(s)
- Anna Berekmeri
- Leeds Institute of Rheumatic and Musculoskeletal Medicine (LIRMM), University of Leeds, Leeds, UK
- National Institute for Health Research (NIHR) Leeds Biomedical Research Centre (BRC), The Leeds Teaching Hospitals, Leeds, UK
| | - Tom Macleod
- Leeds Institute of Rheumatic and Musculoskeletal Medicine (LIRMM), University of Leeds, Leeds, UK
| | - Isabel Hyde
- Leeds Institute of Rheumatic and Musculoskeletal Medicine (LIRMM), University of Leeds, Leeds, UK
| | - Gregor Jan Ojak
- Department of Dermatology, University Medical Centre, Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Caroline Mann
- Department of Dermatology, University Medical Centre, Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Daniela Kramer
- Department of Dermatology, University Medical Centre, Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Martin Stacey
- School of Molecular and Cellular Biology, University of Leeds, Leeds, UK
| | - Miriam Wittmann
- Department of Dermatology, University Medical Centre, Johannes Gutenberg-University Mainz, Mainz, Germany
| |
Collapse
|
39
|
Tu Y, An R, Gu H, Li N, Yan H, Liu HY, He L. The water extracts from the oil cakes of Prinsepia utilis repair the epidermal barrier via up-regulating Corneocyte Envelope-proteins, lipid synthases, and tight junction proteins. JOURNAL OF ETHNOPHARMACOLOGY 2024; 330:118194. [PMID: 38641077 DOI: 10.1016/j.jep.2024.118194] [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: 10/21/2023] [Revised: 04/09/2024] [Accepted: 04/11/2024] [Indexed: 04/21/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Prinsepia utilis Royle, native to the Himalayan region, has a long history of use in traditional medicine for its heat-clearing, detoxification, anti-inflammatory, and analgesic properties. Oils extracted from P. utilis seeds are also used in cooking and cosmetics. With the increasing market demand, this extraction process generates substantial industrial biowastes. Recent studies have found many health benefits with using aqueous extracts of these biowastes, which are also rich in polysaccharides. However, there is limited research related to the reparative effects of the water extracts of P. utilis oil cakes (WEPUOC) on disruptions of the skin barrier function. AIM OF THE STUDY This study aimed to evaluate the reparative efficacy of WEPUOC in both acute and chronic epidermal permeability barrier disruptions. Furthermore, the study sought to explore the underlying mechanisms involved in repairing the epidermal permeability barrier. MATERIALS AND METHODS Mouse models with induced epidermal disruptions, employing tape-stripping (TS) and acetone wiping (AC) methods, were used. The subsequent application of WEPUOC (100 mg/mL) was evaluated through various assessments, with a focus on the upregulation of mRNA and protein expression of Corneocyte Envelope (CE) related proteins, lipid synthase-associated proteins, and tight junction proteins. RESULTS The polysaccharide was the major phytochemicals of WEPUOC and its content was determined as 32.2% by the anthranone-sulfuric acid colorimetric method. WEPUOC significantly reduced transepidermal water loss (TEWL) and improved the damaged epidermal barrier in the model group. Mechanistically, these effects were associated with heightened expression levels of key proteins such as FLG (filaggrin), INV (involucrin), LOR (loricrin), SPT, FASN, HMGCR, Claudins-1, Claudins-5, and ZO-1. CONCLUSIONS WEPUOC, obtained from the oil cakes of P. utilis, is rich in polysaccharides and exhibits pronounced efficacy in repairing disrupted epidermal barriers through increased expression of critical proteins involved in barrier integrity. Our findings underscore the potential of P. utilis wastes in developing natural cosmetic prototypes for the treatment of diseases characterized by damaged skin barriers, including atopic dermatitis and psoriasis.
Collapse
Affiliation(s)
- Ying Tu
- Department of Dermatology, First Affiliated Hospital of Kunming Medical University, Kunming, 650032, China
| | - Ran An
- Department of Dermatology, First Affiliated Hospital of Kunming Medical University, Kunming, 650032, China
| | - Hua Gu
- Department of Dermatology, First Affiliated Hospital of Kunming Medical University, Kunming, 650032, China
| | - Na Li
- Department of Dermatology, First Affiliated Hospital of Kunming Medical University, Kunming, 650032, China
| | - Huan Yan
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650201, China; Yunnan Characteristic Plant Extraction Laboratory, Kunming, 650106, China
| | - Hai-Yang Liu
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650201, China; Yunnan Characteristic Plant Extraction Laboratory, Kunming, 650106, China.
| | - Li He
- Department of Dermatology, First Affiliated Hospital of Kunming Medical University, Kunming, 650032, China.
| |
Collapse
|
40
|
Ozdemir C, Kucuksezer UC, Ogulur I, Pat Y, Yazici D, Ardicli S, Akdis M, Nadeau K, Akdis CA. Lifestyle Changes and Industrialization in the Development of Allergic Diseases. Curr Allergy Asthma Rep 2024; 24:331-345. [PMID: 38884832 PMCID: PMC11233349 DOI: 10.1007/s11882-024-01149-7] [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] [Accepted: 05/02/2024] [Indexed: 06/18/2024]
Abstract
PURPOSE OF REVIEW Modernization and Westernization in industrialized and developing nations is associated with a substantial increase in chronic noncommunicable diseases. This transformation has far-reaching effects on lifestyles, impacting areas such as economics, politics, social life, and culture, all of which, in turn, have diverse influences on public health. Loss of contact with nature, alternations in the microbiota, processed food consumption, exposure to environmental pollutants including chemicals, increased stress and decreased physical activity jointly result in increases in the frequency of inflammatory disorders including allergies and many autoimmune and neuropsychiatric diseases. This review aims to investigate the relationship between Western lifestyle and inflammatory disorders. RECENT FINDINGS Several hypotheses have been put forth trying to explain the observed increases in these diseases, such as 'Hygiene Hypothesis', 'Old Friends', and 'Biodiversity and Dysbiosis'. The recently introduced 'Epithelial Barrier Theory' incorporates these former hypotheses and suggests that toxic substances in cleaning agents, laundry and dishwasher detergents, shampoos, toothpastes, as well as microplastic, packaged food and air pollution damage the epithelium of our skin, lungs and gastrointestinal system. Epithelial barrier disruption leads to decreased biodiversity of the microbiome and the development of opportunistic pathogen colonization, which upon interaction with the immune system, initiates local and systemic inflammation. Gaining a deeper comprehension of the interplay between the environment, microbiome and the immune system provides the data to assist with legally regulating the usage of toxic substances, to enable nontoxic alternatives and to mitigate these environmental challenges essential for fostering a harmonious and healthy global environment.
Collapse
Affiliation(s)
- Cevdet Ozdemir
- Institute of Child Health, Department of Pediatric Basic Sciences, Istanbul University, Istanbul, Türkiye
- Istanbul Faculty of Medicine, Department of Pediatrics, Division of Pediatric Allergy and Immunology, Istanbul University, Istanbul, Türkiye
| | - Umut Can Kucuksezer
- Department of Immunology, Aziz Sancar Institute of Experimental Medicine, Istanbul University, Istanbul, Türkiye
| | - Ismail Ogulur
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Yagiz Pat
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Duygu Yazici
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Sena Ardicli
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
- Department of Genetics, Faculty of Veterinary Medicine, Bursa Uludag University, Bursa, Türkiye
| | - Mubeccel Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Kari Nadeau
- Department of Environmental Studies, Harvard T.H. Chan School of Public Health, Cambridge, MA, USA
| | - Cezmi A Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland.
| |
Collapse
|
41
|
Grieco T, Moliterni E, Paolino G, Chello C, Sernicola A, Egan CG, Nannipieri F, Battaglia S, Accoto M, Tirotta E, Trasciatti S, Bonaretti S, Nencioni S, Biasci E, Pellacani G, Calvieri S. Association between Vitamin D Receptor Polymorphisms, Tight Junction Proteins and Clinical Features of Adult Patients with Atopic Dermatitis. Dermatol Pract Concept 2024; 14:dpc.1403a214. [PMID: 39122483 PMCID: PMC11313695 DOI: 10.5826/dpc.1403a214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/30/2024] [Indexed: 08/12/2024] Open
Abstract
INTRODUCTION Few studies have explored the intricate connections between vitamin D receptor (VDR) gene polymorphisms, VDR, tight junction (TJ) protein expression and clinical features of atopic dermatitis (AD). METHODS From 43 adult AD patients, VDR polymorphisms were genotyped from peripheral blood samples using polymerase chain reaction-restriction fragment length polymorphism. VDR, occludin, claudin-1 and ZO-1 protein expression from skin lesion biopsies were assessed by immunohistochemistry. RESULTS The A1012G heterozygous VDR polymorphism exhibited a lower odds ratio (OR) for juvenile AD onset (OR: 0.046, 95% CI 0.004-0.51, p=0.012). In contrast, the presence of ≥2 homozygous VDR polymorphisms were significantly associated with positive skin prick test (SPT) (10/20, 50%) vs. negative SPT (1/23, 4.3%; p=0.0003). The most highly expressed TJ proteins in lesions of AD patients were claudin-1 and zonulin-1 (ZO-1), while VDR and occludin were less prevalent. A significant correlation was observed between ZO-1 expression and a body mass index ≥30 kg/m2 (OR: 12.1, 95% CI 1.06-137.9, p=0.045). Claudin-1 expression was associated with a positive SPT (OR: 8.23, 95% CI 1.04-65.5, p=0.046) and serum 25(OH)D levels were negatively correlated with ZO-1 expression (rho= -0.43, p=0.0058). CONCLUSION This study provides novel insights into the relationship between VDR gene polymorphisms, VDR, TJ protein expression, and clinical features in adult AD patients, highlighting a significant role of vitamin D in the pathophysiology of this disease.
Collapse
Affiliation(s)
- Teresa Grieco
- Dermatology Clinic, Department of Clinical Internal, Anesthesiological and Cardiovascular Sciences, Sapienza University of Rome, Rome, Italy
| | - Elisa Moliterni
- Dermatology Clinic, Department of Clinical Internal, Anesthesiological and Cardiovascular Sciences, Sapienza University of Rome, Rome, Italy
| | - Giovanni Paolino
- Unit of Dermatology and Cosmetology, IRCCS University Vita-Salute San Raffaele, Milan, Italy
| | - Camilla Chello
- Dermatology Clinic, Department of Clinical Internal, Anesthesiological and Cardiovascular Sciences, Sapienza University of Rome, Rome, Italy
| | - Alvise Sernicola
- Dermatology Clinic, Department of Clinical Internal, Anesthesiological and Cardiovascular Sciences, Sapienza University of Rome, Rome, Italy
| | | | | | | | | | | | | | | | | | | | - Giovanni Pellacani
- Dermatology Clinic, Department of Clinical Internal, Anesthesiological and Cardiovascular Sciences, Sapienza University of Rome, Rome, Italy
| | - Stefano Calvieri
- Dermatology Clinic, Department of Clinical Internal, Anesthesiological and Cardiovascular Sciences, Sapienza University of Rome, Rome, Italy
| |
Collapse
|
42
|
Lin S, Liu X, Jiang J, Ge W, Zhang Y, Li F, Tao Q, Liu S, Li M, Chen H. The involvement of keratinocytes in pruritus of chronic inflammatory dermatosis. Exp Dermatol 2024; 33:e15142. [PMID: 39032085 DOI: 10.1111/exd.15142] [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: 07/24/2023] [Revised: 07/02/2024] [Accepted: 07/09/2024] [Indexed: 07/22/2024]
Abstract
Frequent itching and incessant scratching are commonly observed in various chronic inflammatory skin conditions, including atopic dermatitis and psoriasis. The persistent and prolonged nature of pruritus can worsen one's quality of life. Keratinocytes (KCs), the predominant cells of the epidermis, have been confirmed to interact with sensory neurons and immune cells and be involved in chronic skin inflammatory diseases associated with pruritus. Initially, KCs and sensory neurons form a unique synapse-like connection within the epidermis, serving as the structural foundation for their interaction. Additionally, several receptors, including toll-like receptors and protease-activated receptor 2, expressed on KCs, become activated in an inflammatory milieu. On the one hand, activated KCs are sources of pro-inflammatory cytokines and neurotrophic factors, such as adenosine triphosphate, thymic stromal lymphopoietin, and nerve growth factor, which directly or indirectly participate in stimulating sensory neurons, thereby contributing to the itch sensations. On the other hand, KCs also function as primary transducers alongside intraepidermal nerve endings, directly initiating pruritic responses. This review summarizes the current literature and highlights the critical role of KCs in the development and persistence of chronic itch in inflammatory skin disorders.
Collapse
Affiliation(s)
- Shiying Lin
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xin Liu
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jian Jiang
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wenqiang Ge
- Department of Neurobiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yinlian Zhang
- Shenzhen Qianhai Shekou Free Trade Zone Hospital, Shenzhen, China
- Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Fei Li
- Department of Dermatology, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen, China
- Department of Dermatology, The 6th Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, China
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen, China
| | - Qingxiao Tao
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Suwen Liu
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Man Li
- Department of Neurobiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hongxiang Chen
- Department of Dermatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Dermatology, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen, China
- Department of Dermatology, The 6th Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, China
| |
Collapse
|
43
|
Wrześniewska M, Wołoszczak J, Świrkosz G, Szyller H, Gomułka K. The Role of the Microbiota in the Pathogenesis and Treatment of Atopic Dermatitis-A Literature Review. Int J Mol Sci 2024; 25:6539. [PMID: 38928245 PMCID: PMC11203945 DOI: 10.3390/ijms25126539] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 06/09/2024] [Accepted: 06/10/2024] [Indexed: 06/28/2024] Open
Abstract
Atopic dermatitis (AD) is a chronic inflammatory skin condition with a high prevalence worldwide. AD pathogenesis is complex and consists of immune system dysregulation and impaired skin barrier, influenced by genetic and environmental factors. The purpose of the review is to show the complex interplay between atopic dermatitis and the microbiota. Human microbiota plays an important role in AD pathogenesis and the course of the disease. Dysbiosis is an important factor contributing to the development of atopic diseases, including atopic dermatitis. The gut microbiota can influence the composition of the skin microbiota, strengthening the skin barrier and regulating the immune response via the involvement of bacterial metabolites, particularly short-chain fatty acids, in signaling pathways of the gut-skin axis. AD can be modulated by antibiotic intake, dietary adjustments, hygiene, and living conditions. One of the promising strategies for modulating the course of AD is probiotics. This review offers a summary of how the microbiota influences the development and treatment of AD, highlighting aspects that warrant additional investigation.
Collapse
Affiliation(s)
- Martyna Wrześniewska
- Student Scientific Group of Internal Medicine and Allergology, Faculty of Medicine, Wroclaw Medical University, 50-368 Wroclaw, Poland; (M.W.); (J.W.); (G.Ś.); (H.S.)
| | - Julia Wołoszczak
- Student Scientific Group of Internal Medicine and Allergology, Faculty of Medicine, Wroclaw Medical University, 50-368 Wroclaw, Poland; (M.W.); (J.W.); (G.Ś.); (H.S.)
| | - Gabriela Świrkosz
- Student Scientific Group of Internal Medicine and Allergology, Faculty of Medicine, Wroclaw Medical University, 50-368 Wroclaw, Poland; (M.W.); (J.W.); (G.Ś.); (H.S.)
| | - Hubert Szyller
- Student Scientific Group of Internal Medicine and Allergology, Faculty of Medicine, Wroclaw Medical University, 50-368 Wroclaw, Poland; (M.W.); (J.W.); (G.Ś.); (H.S.)
| | - Krzysztof Gomułka
- Clinical Department of Internal Medicine, Pneumology and Allergology, Faculty of Medicine, Wroclaw Medical University, 50-368 Wroclaw, Poland
| |
Collapse
|
44
|
Wong TK, Choi YG, Li PH, Chow BKC, Kumar M. MRGPRX2 antagonist GE1111 attenuated DNFB-induced atopic dermatitis in mice by reducing inflammatory cytokines and restoring skin integrity. Front Immunol 2024; 15:1406438. [PMID: 38817611 PMCID: PMC11137259 DOI: 10.3389/fimmu.2024.1406438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Accepted: 04/26/2024] [Indexed: 06/01/2024] Open
Abstract
Introduction Atopic dermatitis (AD) is a chronic inflammatory skin disorder characterised by itching, erythema, and epidermal barrier dysfunction. The pathogenesis of AD is complex and multifactorial; however,mast cell (MC) activation has been reported to be one of the crucial mechanisms in the pathogenesis of AD. The MC receptor Mas related G protein-coupled receptor-X2 (MRGPRX2) has been identified as a prominent alternative receptor to the IgE receptor in causing MC activation and the subsequent release of inflammatory mediators. The current study aimed to evaluate the therapeutic effect of a novel small molecule MRGPRX2 antagonist GE1111 in AD using in vitro and in vivo approaches. Methods We developed an in vitro cell culture disease model by using LAD-2 MC, HaCaT keratinocytes and RAW 264.7 macrophage cell lines. We challenged keratinocytes and macrophage cells with CST-14 treated MC supernatant in the presence and absence of GE1111 and measured the expression of tight junction protein claudin 1, inflammatory cytokines and macrophage phagocytosis activity through immunohistochemistry, western blotting, RT-qPCR and fluorescence imaging techniques. In addition to this, we developed a DFNB-induced AD model in mice and evaluated the protective effect and underlying mechanism of GE1111. Results and Discussion Our in vitro findings demonstrated a potential therapeutic effect of GE1111, which inhibits the expression of TSLP, IL-13, MCP-1, TNF-a, and IL-1ß in MC and keratinocytes. In addition to this, GE1111 was able to preserve the expression of claudin 1 in keratinocytes and the phagocytotic activity of macrophage cells. The in vivo results demonstrated that GE1111 treatment significantly reduced phenotypic changes associated with AD (skin thickening, scaling, erythema and epidermal thickness). Furthermore, immunohistochemical analysis demonstrated that GE1111 treatment preserved the expression of the tight junction protein Involucrin and reduced the expression of the inflammatory mediator periostin in the mouse model of AD. These findings were supported by gene and protein expression analysis, where GE1111 treatment reduced the expression of TSLP, IL-13, and IL-1ß, as well as downstream signalling pathways of MRGPRX2 in AD skin lesions. In conclusion, our findings provide compelling in vitro and in vivo evidence supporting the contribution of MRGPRX2-MC interaction with keratinocytes and macrophages in the pathogenesis of AD.
Collapse
Affiliation(s)
- Trevor K. Wong
- School of Biological Sciences, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
- Faculty of Health Sciences, McMaster University, Hamliton, ON, Canada
| | - Ye Gi Choi
- School of Biological Sciences, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Philip H. Li
- Division of Rheumatology and Clinical Immunology, Department of Medicine, Queen Mary Hospital, University of Hong Kong, Hong Kong SAR, China
| | - Billy K. C. Chow
- School of Biological Sciences, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Mukesh Kumar
- School of Biological Sciences, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| |
Collapse
|
45
|
Arnold KA, Moran MC, Shi H, van Vlijmen-Willems IMJJ, Rodijk-Olthuis D, Smits JPH, Brewer MG. CLDN1 knock out keratinocytes as a model to investigate multiple skin disorders. Exp Dermatol 2024; 33:e15084. [PMID: 38711223 DOI: 10.1111/exd.15084] [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: 02/20/2023] [Revised: 02/02/2024] [Accepted: 03/05/2024] [Indexed: 05/08/2024]
Abstract
The transmembrane protein claudin-1 is critical for formation of the epidermal barrier structure called tight junctions (TJ) and has been shown to be important in multiple disease states. These include neonatal ichthyosis and sclerosing cholangitis syndrome, atopic dermatitis and various viral infections. To develop a model to investigate the role of claudin-1 in different disease settings, we used CRISPR/Cas9 to generate human immortalized keratinocyte (KC) lines lacking claudin-1 (CLDN1 KO). We then determined whether loss of claudin-1 expression affects epidermal barrier formation/function and KC differentiation/stratification. The absence of claudin-1 resulted in significantly reduced barrier function in both monolayer and organotypic cultures. CLDN1 KO cells demonstrated decreases in gene transcripts encoding the barrier protein filaggrin and the differentiation marker cytokeratin-10. Marked morphological differences were also observed in CLDN1 KO organotypic cultures including diminished stratification and reduced formation of the stratum granulosum. We also detected increased proliferative KC in the basale layer of CLDN1 KO organotypic cultures. These results further support the role of claudin-1 in epidermal barrier and suggest an additional role of this protein in appropriate stratification of the epidermis.
Collapse
Affiliation(s)
- Kimberly A Arnold
- Department of Dermatology, University of Rochester Medical Center, Rochester, New York, USA
| | - Mary C Moran
- Department of Dermatology, University of Rochester Medical Center, Rochester, New York, USA
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, New York, USA
| | - Huishan Shi
- Department of Dermatology, University of Rochester Medical Center, Rochester, New York, USA
| | - Ivonne M J J van Vlijmen-Willems
- Department of Dermatology, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences (RIMLS), Nijmegen, The Netherlands
| | - Diana Rodijk-Olthuis
- Department of Dermatology, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences (RIMLS), Nijmegen, The Netherlands
| | - Jos P H Smits
- Department of Dermatology, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences (RIMLS), Nijmegen, The Netherlands
- Department of Dermatology, University Hospital Düsseldorf, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany
| | - Matthew G Brewer
- Department of Dermatology, University of Rochester Medical Center, Rochester, New York, USA
| |
Collapse
|
46
|
Napolitano M, Potestio L, Nocerino M, Patruno C. Considerations for managing elderly patients with atopic dermatitis. Expert Rev Clin Immunol 2024; 20:501-511. [PMID: 38193289 DOI: 10.1080/1744666x.2024.2301967] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2023] [Accepted: 12/22/2023] [Indexed: 01/10/2024]
Abstract
INTRODUCTION Atopic dermatitis (AD) diagnosis in elderly is challenging, due to its clinical polymorphism and the lack of diagnostic biomarkers. Moreover, the chronicity of the disease and the complex pathogenetic mechanism, make elderly AD management challenging. AREAS COVERED A narrative review of the current literature was performed using the PubMed, Medline, Embase, and Cochrane Skin databases, by researching the following terms: 'atopic dermatitis,' 'clinical phenotypes,' 'eczema,' 'elderly patients,' 'elderly type atopic dermatitis,' 'eczema clinical presentation.' The aim was to report the current knowledge on pathogenesis, clinical presentation, and treatment options of elderly AD. EXPERT OPINION Elderly type AD has recently been identified as a separate entity, with an increasing prevalence. With aging, both immunosenescence and barrier alterations can cause or modify AD presentation. Moreover, a chronic proinflammatory state (so-called 'inflammaging') is often present in elderly subjects. Older patients with AD may present with peculiar immunophenotypic profile, making AD diagnosis challenging. Similarly, the chronicity of the disease and the complex pathogenetic mechanism, make AD management a challenge. Indeed, systemic therapies for AD are often contraindicated or not tolerated and the management of elderly type AD is often burdened with numerous difficulties, leading to undertreated disease. Even if dupilumab and tralokinumab represent a valuable therapeutic weapon, more data on safety of JAK inhibitors are required.
Collapse
Affiliation(s)
- Maddalena Napolitano
- Section of Dermatology, Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Luca Potestio
- Section of Dermatology, Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Mariateresa Nocerino
- Section of Dermatology, Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Cataldo Patruno
- Department of Health Sciences, University Magna Graecia of Catanzaro, Catanzaro, Italy
| |
Collapse
|
47
|
Zhang Y, Lu Q. Immune cells in skin inflammation, wound healing, and skin cancer. J Leukoc Biol 2024; 115:852-865. [PMID: 37718697 DOI: 10.1093/jleuko/qiad107] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2023] [Revised: 08/22/2023] [Accepted: 09/05/2023] [Indexed: 09/19/2023] Open
Abstract
Given the self-evident importance of cutaneous immunity in the maintenance of body-surface homeostasis, disturbance of the steady-state skin is inextricably intertwined with dysfunction in cutaneous immunity. It is often overlooked by people that skin, well-known as a solid physical barrier, is also a strong immunological barrier, considering the abundant presence of immune cells including lymphocytes, granulocytes, dendritic cells, and macrophages. What's more, humoral immune components including cytokines, immunoglobulins, and antimicrobial peptides are also rich in the skin. This review centers on skin inflammation (acute and chronic, infection and aseptic inflammation), wound healing, and skin cancer to elucidate the elaborate network of immune cells in skin diseases.
Collapse
Affiliation(s)
- Yuhan Zhang
- Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Jiangwangmiao Street No. 12, Xuanwu, Nanjing 210042, China
- Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Jiangwangmiao Street No. 12, Xuanwu, Nanjing 210042, China
- Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing 210042, China
| | - Qianjin Lu
- Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Jiangwangmiao Street No. 12, Xuanwu, Nanjing 210042, China
- Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Jiangwangmiao Street No. 12, Xuanwu, Nanjing 210042, China
- Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing 210042, China
| |
Collapse
|
48
|
Morgenstern AR, Peterson LF, Arnold KA, Brewer MG. Differentiation of keratinocytes or exposure to type 2 cytokines diminishes S. aureus internalization. mSphere 2024; 9:e0068523. [PMID: 38501828 PMCID: PMC11036805 DOI: 10.1128/msphere.00685-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Accepted: 02/26/2024] [Indexed: 03/20/2024] Open
Abstract
Staphylococcus aureus is a leading cause of skin and soft tissue infections. Colonization by this bacterium is increased in individuals with chronic cutaneous diseases such as atopic dermatitis, psoriasis, and bullous pemphigoid. The greater abundance of S. aureus on the skin of subjects with atopic dermatitis in particular has been linked to recurrent cutaneous infections. The primary cell type of the epidermal layer of the skin is the keratinocyte, and it is thought that S. aureus internalized in keratinocytes associates with an increased incidence of skin infections. This study addresses whether keratinocyte differentiation and/or inflammation, two important characteristics altered in cutaneous diseases, influence bacterial internalization. To do this, S. aureus internalization was measured in immortalized and primary keratinocytes that were differentiated using high Ca2+-containing media and/or exposed to cytokines characteristic of atopic dermatitis (IL-4 and IL-13) or psoriasis (IL-17A and IL-22) skin. Our results indicate that S. aureus internalization is uniquely decreased upon keratinocyte differentiation, since this was not observed with another skin-resident bacterium, S. epidermidis. Additionally, treatment with IL-4 + IL-13 diminished bacterial internalization. We interpret this decrease as a mechanism of keratinocyte-based bacterial killing since a similar number of bacterial genomes were detected in cytokine-treated cells, but less viable internalized S. aureus was recovered. Finally, of the receptors reported for S. aureus binding/internalizing into keratinocytes, expression of the α5 component of the α5β1 integrin was in greatest accordance with the number of internalized bacteria in the context of keratinocyte differentiation.IMPORTANCEIndividuals with chronic cutaneous diseases demonstrate heightened susceptibility for severe and recurrent infections from Staphylococcus aureus. What drives this altered susceptibility remains poorly understood. Previous publications have detected S. aureus as deep as the dermal layer of skin in subjects with atopic dermatitis, suggesting that the cutaneous environment of this disease enables deeper bacterial infiltration than occurs in healthy individuals. This observation indicates that S. aureus has greater opportunity to interact with multiple skin cell types in individuals with chronic inflammatory skin diseases. Identifying the characteristics of the skin that influence bacterial internalization, a common method to establish reservoirs and evade the immune response, is critical for our understanding of S. aureus pathogenesis. The significance of this research is the novel identification of epidermal characteristics that influence S. aureus internalization. With this knowledge, methods can be developed to identify patient populations at greater risk for cutaneous infections.
Collapse
Affiliation(s)
| | - Liam F. Peterson
- Department of Pathology & Laboratory Medicine, University of Rochester, Rochester, New York, USA
| | - Kimberly A. Arnold
- Department of Dermatology, University of Rochester, Rochester, New York, USA
| | - Matthew G. Brewer
- Department of Dermatology, University of Rochester, Rochester, New York, USA
| |
Collapse
|
49
|
Afshari M, Kolackova M, Rosecka M, Čelakovská J, Krejsek J. Unraveling the skin; a comprehensive review of atopic dermatitis, current understanding, and approaches. Front Immunol 2024; 15:1361005. [PMID: 38500882 PMCID: PMC10944924 DOI: 10.3389/fimmu.2024.1361005] [Citation(s) in RCA: 26] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2023] [Accepted: 02/14/2024] [Indexed: 03/20/2024] Open
Abstract
Atopic dermatitis, also known as atopic eczema, is a chronic inflammatory skin disease characterized by red pruritic skin lesions, xerosis, ichthyosis, and skin pain. Among the social impacts of atopic dermatitis are difficulties and detachment in relationships and social stigmatization. Additionally, atopic dermatitis is known to cause sleep disturbance, anxiety, hyperactivity, and depression. Although the pathological process behind atopic dermatitis is not fully known, it appears to be a combination of epidermal barrier dysfunction and immune dysregulation. Skin is the largest organ of the human body which acts as a mechanical barrier to toxins and UV light and a natural barrier against water loss. Both functions face significant challenges due to atopic dermatitis. The list of factors that can potentially trigger or contribute to atopic dermatitis is extensive, ranging from genetic factors, family history, dietary choices, immune triggers, and environmental factors. Consequently, prevention, early clinical diagnosis, and effective treatment may be the only resolutions to combat this burdensome disease. Ensuring safe and targeted drug delivery to the skin layers, without reaching the systemic circulation is a promising option raised by nano-delivery systems in dermatology. In this review, we explored the current understanding and approaches of atopic dermatitis and outlined a range of the most recent therapeutics and dosage forms brought by nanotechnology. This review was conducted using PubMed, Google Scholar, and ScienceDirect databases.
Collapse
Affiliation(s)
- Moeina Afshari
- Department of Clinical Immunology and Allergy, Faculty Hospital and Medical Faculty of Charles University, Hradec Králové, Czechia
| | - Martina Kolackova
- Department of Clinical Immunology and Allergy, Faculty Hospital and Medical Faculty of Charles University, Hradec Králové, Czechia
| | - Michaela Rosecka
- Department of Clinical Immunology and Allergy, Faculty Hospital and Medical Faculty of Charles University, Hradec Králové, Czechia
| | - Jarmila Čelakovská
- Department of Dermatology and Venereology, Faculty Hospital and Medical Faculty of Charles University, Hradec Králové, Czechia
| | - Jan Krejsek
- Department of Clinical Immunology and Allergy, Faculty Hospital and Medical Faculty of Charles University, Hradec Králové, Czechia
| |
Collapse
|
50
|
Chen HW, Liu FC, Kuo HM, Tang SH, Niu GH, Zhang MM, Tsou LK, Sung PJ, Wen ZH. Immunomodulatory and anti-angiogenesis effects of excavatolide B and its derivatives in alleviating atopic dermatitis. Biomed Pharmacother 2024; 172:116279. [PMID: 38368838 DOI: 10.1016/j.biopha.2024.116279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 02/03/2024] [Accepted: 02/13/2024] [Indexed: 02/20/2024] Open
Abstract
Atopic dermatitis (AD) is a chronic inflammatory skin condition primarily driven by T helper 2 (Th2) cytokines, resulting in skin barrier defects, angiogenesis, and inflammatory responses. The marine natural product excavatolide B (EXCB), isolated from the Formosan Gorgonian coral Briareum stechei, exhibits anti-inflammatory and analgesic properties. To enhance solubility, EXCB is chemically modified into the derivatives EXCB-61 salt and EXCB-79. The study aims to investigate the therapeutic effects of these compounds on dinitrochlorbenzene (DNCB)-induced skin damage and to elucidate the underlying anti-inflammatory and anti-angiogenesis mechanism. In vitro, using lipopolysaccharide (LPS)-induced RAW 264.7 cells, all compounds at 10 μM significantly inhibited expression of inflammatory proteins (inducible nitric oxide synthase and cyclooxygenase-2), vascular endothelial growth factor (VEGF), and cytokines (interleukin (IL)-1β, IL-6, and IL-17A). In vivo, topical application of these compounds on DNCB-induced AD mice alleviated skin symptoms, reduced serum levels of IgE, IL-4, IL-13, IL-17, and interferon-γ, and moderated histological phenomena such as hyperplasia, inflammatory cell infiltration, and angiogenesis. The three compounds restored the expression of skin barrier-related proteins (loricrin, filaggrin, and claudin-1) and reduced the expression of angiogenesis-related proteins (VEGF and platelet endothelial cell adhesion molecule-CD31) in the tissues. This is the first study to indicate that EXCB, EXCB-61 salt, and EXCB-79 can treat AD disease by reducing inflammation and angiogenesis. Hence, they may be considered potential candidates for the development of new drugs for AD.
Collapse
Affiliation(s)
- Hsiu-Wen Chen
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Kaohsiung 80424, Taiwan
| | - Feng-Cheng Liu
- Division of Rheumatology/Immunology and Allergy, Department of Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei 114202, Taiwan
| | - Hsiao-Mei Kuo
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Kaohsiung 80424, Taiwan; Department of Neurosurgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University, College of Medicine, Kaohsiung 83301, Taiwan
| | - Shih-Hsuan Tang
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Kaohsiung 80424, Taiwan
| | - Guang-Hao Niu
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli, Zhunan 35053, Taiwan
| | - Mingzi M Zhang
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Miaoli, Zhunan 35053, Taiwan
| | - Lun Kelvin Tsou
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli, Zhunan 35053, Taiwan.
| | - Ping-Jyun Sung
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Kaohsiung 80424, Taiwan; National Museum of Marine Biology and Aquarium, Pingtung 94450, Taiwan.
| | - Zhi-Hong Wen
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Kaohsiung 80424, Taiwan; Institute of Biotechnology and Pharmaceutical Research, National Sun Yat-sen University, Kaohsiung 80424, Taiwan.
| |
Collapse
|