1
|
Chen Z, Yang Y, Wang X, Xia L, Wang W, Wu X, Gao Z. Keloids and inflammation: the crucial role of IL-33 in epidermal changes. Front Immunol 2025; 16:1514618. [PMID: 40230853 PMCID: PMC11994421 DOI: 10.3389/fimmu.2025.1514618] [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: 10/21/2024] [Accepted: 03/14/2025] [Indexed: 04/16/2025] Open
Abstract
Introduction Keloids are benign fibroproliferative disorders characterized by excessive collagen deposition and inflammation that extend beyond the original wound boundaries. IL-33 is an alarmin cytokine released upon cellular damage or stress. Dysregulation of IL-33 in epidermal keratinocytes compromises the skin barrier and triggers chronic inflammation. Method In this study, we first noticed an increased expression of IL-33 in the keratinocytes of keloid epidermis through histological staining. Then, an increased expression of IL-33 receptor (ST2) in the lymphocytes infiltrating the superficial dermis of keloid scars were identified through histological staining and flow cytometry analysis. The IFN-γ-IL-33 loop between lymphocytes and keratinocytes were further revealed by flow cytometry and Western blotting analysis. The abnormal keratinocyte differentiation in epiderm is mediated by IFN-γ-IL-33 loop were confirmed by in vitro studies in HaCaT cells via Western blotting analysis and immunofluorescence staining. Finally, the IFN-γ-IL-33 loop were also verified in cocultured peripheral blood mononuclear cells and HaCaT through ELISA analysis. Results Our results demonstrate that IL-33 levels are significantly elevated in the epidermis of keloid tissues, where it functions as an alarmin, promoting a chronic inflammatory response. We further reveal a feedback loop between IL-33 and interferon-gamma (IFN-γ), whereby IL-33 induces IFN-g production in lymphocytes, which in turn stimulates keratinocytes to produce more IL-33. This loop contributes to impaired keratinocyte differentiation and skin barrier dysfunction, exacerbating the inflammatory environment. Discussion By elucidating the role of the IL-33/ST2 axis in keloid formation, this research provides valuable insights into potential therapeutic targets for managing this challenging condition.
Collapse
Affiliation(s)
| | | | | | | | | | - XiaoLi Wu
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhen Gao
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
2
|
Meng J, Xiao H, Xu F, She X, Liu C, Canonica GW. Systemic barrier dysfunction in type 2 inflammation diseases: perspective in the skin, airways, and gastrointestinal tract. Immunol Res 2025; 73:60. [PMID: 40069459 PMCID: PMC11897119 DOI: 10.1007/s12026-025-09606-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Accepted: 02/13/2025] [Indexed: 03/15/2025]
Abstract
The epithelial barrier in different organs is the first line of defense against environmental insults and allergens, with type 2 immunity serving as a protective function. Genetic factors, and biological and chemical insults from the surrounding environment altered regulate epithelial homeostasis through disruption of epithelial tight junction proteins or dilated intercellular spaces. Recent studies suggest that epithelial barrier dysfunction contributes to pathologic alteration in diseases with type 2 immune dysregulation including (but not limited to) atopic dermatitis, prurigo nodularis, asthma, chronic rhinosinusitis with nasal polyps, and eosinophilic esophagitis. In this review, we summarized current understanding of dysfunction of barrier and its interaction with type 2 inflammation across different organs, and discussed the role of epithelial barrier disruption in the pathogenesis of type 2 inflammation. In addition, recent progresses of emerging barrier restorative therapies are reviewed.
Collapse
Affiliation(s)
- Juan Meng
- Department of Allergy, West China Hospital, Sichuan University, Chengdu, China
- Department of Otorhinolaryngology, West China Hospital, Sichuan University, Chengdu, China
| | - Hao Xiao
- Department of Allergy, West China Hospital, Sichuan University, Chengdu, China
| | - Feng Xu
- Department of Allergy, West China Hospital, Sichuan University, Chengdu, China
| | - Xueke She
- Sanofi China Investment Co., Ltd. Shanghai Branch, Shanghai, 200000, P.R. China
| | - Chuntao Liu
- Department of Allergy, West China Hospital, Sichuan University, Chengdu, China.
- Department of Respiratory Medicine, West China Hospital, Sichuan University, Chengdu, China.
| | - Giorgio Walter Canonica
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, Pieve Emanuele, Milan, Italy
- Asthma & Allergy Unit, IRCCS Humanitas Research Hospital, Via Manzoni 56, Rozzano, Milan, Italy
| |
Collapse
|
3
|
Atamas SP, Lockatell V, Todd NW, Papadimitriou JC, Rus V, Lugkey KN, Vogel SN, Toshchakov VY, Luzina IG. Therapeutic targeting of full-length interleukin-33 protein levels with cell-permeable decoy peptides attenuates fibrosis in the bleomycin model in vivo. J Pharmacol Exp Ther 2025; 392:100008. [PMID: 39893007 DOI: 10.1124/jpet.123.002050] [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: 12/01/2023] [Revised: 05/08/2024] [Accepted: 05/20/2024] [Indexed: 06/12/2024] Open
Abstract
Interleukin (IL)-33 has been shown to centrally regulate, among other processes, inflammation and fibrosis. Both intracellular full-length (FLIL33) precursor and extracellular mature cytokine (MIL33) forms exert such regulation, albeit differentially. Drug development efforts to target the IL-33 pathway have focused mostly on MIL33 and its specific cell-surface receptor, ST2, with limited attempts to negotiate the pathophysiological contributions from FLIL33. Furthermore, even a successful strategy for targeting MIL33 effects would arguably benefit from a simultaneous attenuation of the levels of FLIL33, which remains the continuous source of MIL33 supply. We therefore sought to develop an approach to depleting FLIL33 protein levels. We previously reported that the steady-state levels of FLIL33 are controlled in part through its proteasomal degradation and that such regulation can be mapped to a segment in the N-terminal portion of FLIL33. We hypothesized that disruption of this regulation would lead to a decrease in FLIL33 levels, thus inducing a beneficial therapeutic effect in an IL-33-dependent pathology. To test this hypothesis, we designed and tested cell-permeable decoy peptides, which mimic the target N-terminal FLIL33 region. We argued that such mimic peptides would compete with FLIL33 for the components of the native FLIL33 production and maintenance molecular machinery. Administered in the therapeutic regimen to bleomycin-challenged mice, the tested cell-permeable decoy peptides alleviated the overall severity of the disease by restoring body weight loss and attenuating accumulation of collagen in the lungs. This proof-of-principle study lays the foundation for future work toward the development of this prospective therapeutic approach. SIGNIFICANCE STATEMENT: An antifibrotic therapeutic approach is proposed and preclinically tested in mice in vivo based on targeting the full-length IL-33 precursor protein. Peptide fusion constructs consisted of a cell-permeable sequence fused with a sequence mimicking an N-terminal segment of IL-33 precursor that is responsible for this protein's stability. Systemic administration of such peptides to mice in either the acute intratracheal or chronic systemic bleomycin challenge models leads to a decrease in the bleomycin-induced elevations of pulmonary IL-33 and collagen.
Collapse
Affiliation(s)
- Sergei P Atamas
- Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland; Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Virginia Lockatell
- Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland
| | - Nevins W Todd
- Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland; Research Service, Baltimore VA Medical Center, Baltimore, Maryland
| | - John C Papadimitriou
- Department of Pathology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Violeta Rus
- Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland
| | - Katerina N Lugkey
- Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland
| | - Stefanie N Vogel
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Vladimir Y Toshchakov
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Irina G Luzina
- Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland; Research Service, Baltimore VA Medical Center, Baltimore, Maryland.
| |
Collapse
|
4
|
Tandon R, Harder I, Stölzl D, Hübenthal M, Sander N, Hartmann J, Suhrkamp I, Fonfara M, Gerdes S, Weidinger S. Tralokinumab Treatment of Atopic Dermatitis Induces a Progressive Transcriptomic Response. J Invest Dermatol 2024:S0022-202X(24)03035-5. [PMID: 39733934 DOI: 10.1016/j.jid.2024.12.005] [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: 07/12/2024] [Revised: 11/05/2024] [Accepted: 12/03/2024] [Indexed: 12/31/2024]
Abstract
Atopic dermatitis is characterized by a complex epidermal barrier deficiency and exaggerated immune responses dominated by type 2 mechanisms with variable contributions of additional immune axes. IL-13 is overexpressed in atopic dermatitis skin and a key driver of both barrier dysfunction and inflammation. In this study, we prospectively studied the effects of IL-13 inhibition with tralokinumab on cutaneous transcriptome profiles using RNA sequencing of biopsies from 16 patients with moderate-to-severe atopic dermatitis obtained at baseline, week 2, and week 16. Tralokinumab therapy induced early and delayed expression changes and progressively shifted the transcriptomic profile of lesional toward nonlesional skin by modulating both genes associated with keratinocyte proliferation and differentiation, itch signaling, and downstream inflammatory responses. At week 16, 751 genes were still significantly dysregulated compared with those in healthy control skin, reinforcing the need for long-term immunomodulatory therapy of moderate-to-severe atopic dermatitis to achieve deep responses. The study was registered with ClinicalTrials.gov (NCT04556461).
Collapse
Affiliation(s)
- Rashmi Tandon
- Department of Dermatology and Allergy, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Inken Harder
- Department of Dermatology and Allergy, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Dora Stölzl
- Department of Dermatology and Allergy, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Matthias Hübenthal
- Department of Dermatology and Allergy, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Nicole Sander
- Department of Dermatology and Allergy, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Jan Hartmann
- Department of Dermatology and Allergy, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Ina Suhrkamp
- Department of Dermatology and Allergy, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Melina Fonfara
- Department of Dermatology and Allergy, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Sascha Gerdes
- Department of Dermatology and Allergy, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Stephan Weidinger
- Department of Dermatology and Allergy, University Hospital Schleswig-Holstein, Kiel, Germany.
| |
Collapse
|
5
|
Dai X, Mizukami Y, Watanabe K, Tsuda T, Shidahara M, Yoshida S, Yatsuzuka K, Shiraishi K, Mori H, Murakami M, Kawakami R, Imamura T, Fujisawa Y, Muto J. Trehalose Prevents IL-4/IL-13-Induced Skin Barrier Impairment by Suppressing IL-33 Expression and Increasing NRF2 Activation in Human Keratinocytes In Vitro. J Invest Dermatol 2024:S0022-202X(24)02175-4. [PMID: 39384017 DOI: 10.1016/j.jid.2024.08.038] [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: 10/07/2023] [Revised: 08/16/2024] [Accepted: 08/25/2024] [Indexed: 10/11/2024]
Abstract
Skin barrier dysfunction initiates or deteriorates various cutaneous problems, such as atopic dermatitis. At high concentrations, the nonreducing disaccharide trehalose (α-d-glucopyranosyl α-d-glucopyranoside) induces a transient senescence-like state in fibroblasts and promotes wound repair. In this study, we investigated the effect of trehalose on normal human keratinocytes and demonstrated its specific role in the skin barrier. RNA-sequencing analysis revealed that trehalose regulates the expression of many skin barrier-associated genes. T helper 2 cytokines IL-4/IL-13 were observed to downregulate several differentiation markers (FLG, loricrin, keratin 1, and keratin 10) and epidermal antimicrobial proteins in monolayer-cultured keratinocytes and living skin equivalents and impaired skin barrier function in living skin equivalents, all of which were significantly upregulated or restored by trehalose. Trehalose inhibited IL-33 expression and reduced nuclear IL-33 levels by activating MAPK/extracellular signal-regulated kinase kinase 5-extracellular signal-regulated kinase 5 and suppressing extracellular signal-regulated kinase kinase 1/2-extracellular signal-regulated kinase pathway. It also increased NRF2 activation to trigger antioxidant enzyme production through JNK, thus neutralizing IL-4/IL-13-mediated oxidative stress. Trehalose prevented IL-4/IL-13-mediated signal transducer and activator of transcription 3/signal transducer and activator of transcription 6 activation and restored IL-4/IL-13-suppressed skin barrier molecules through IL-33 downregulation and NRF2 activation. This study demonstrated that trehalose may play a role in skin barrier repair in atopic dermatitis.
Collapse
Affiliation(s)
- Xiuju Dai
- Department of Dermatology, Ehime University Graduate School of Medicine, Ehime, Japan; Department of Dermatology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiaotong University, Shanghai, China
| | - Yoichi Mizukami
- Institute of Gene Research, Yamaguchi University Science Research Center, Yamaguchi, Japan
| | - Kenji Watanabe
- Institute of Gene Research, Yamaguchi University Science Research Center, Yamaguchi, Japan
| | - Teruko Tsuda
- Department of Dermatology, Ehime University Graduate School of Medicine, Ehime, Japan
| | - Mutsumi Shidahara
- Department of Dermatology, Ehime University Graduate School of Medicine, Ehime, Japan
| | - Satoshi Yoshida
- Department of Dermatology, Ehime University Graduate School of Medicine, Ehime, Japan
| | - Kazuki Yatsuzuka
- Department of Dermatology, Ehime University Graduate School of Medicine, Ehime, Japan
| | - Ken Shiraishi
- Department of Dermatology, Ehime University Graduate School of Medicine, Ehime, Japan
| | - Hideki Mori
- Department of Dermatology, Ehime University Graduate School of Medicine, Ehime, Japan
| | - Masamoto Murakami
- Department of Dermatology, Ehime University Graduate School of Medicine, Ehime, Japan
| | - Ryosuke Kawakami
- Department of Molecular Medicine for Pathogenesis, Ehime University Graduate School of Medicine, Ehime, Japan
| | - Takeshi Imamura
- Department of Molecular Medicine for Pathogenesis, Ehime University Graduate School of Medicine, Ehime, Japan; Translational Research Center, Ehime University Hospital, Ehime, Japan
| | - Yasuhiro Fujisawa
- Department of Dermatology, Ehime University Graduate School of Medicine, Ehime, Japan
| | - Jun Muto
- Department of Dermatology, Ehime University Graduate School of Medicine, Ehime, Japan.
| |
Collapse
|
6
|
Lu B, Zhao S, Zhang J, Zhan J, Zhang J, Liu Z, Zhang J. Anti-inflammatory and antioxidant effects on skin based on supramolecular hyaluronic acid-ectoin. J Mater Chem B 2024; 12:8408-8419. [PMID: 39086221 DOI: 10.1039/d4tb00459k] [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: 08/02/2024]
Abstract
We addressed the damage caused by internal and external factors on the skin, as well as the aging phenomenon caused by delayed repair after damage. We prepared supramolecular hyaluronic acid-ectoin (HA-ECT) by combining theoretical calculations and experimental research, using intermolecular forces between hyaluronic acid and ectoin. This supramolecule has good stability, safety, and skin permeability and can penetrate the stratum corneum of the skin, reaching the epidermis and dermis of the skin. Compared with ectoin, the permeability of the supramolecule HA-ECT was 3.39-fold higher. Supramolecular HA-ECT can promote the proliferation of keratinocytes and fibroblasts, significantly increase the content of type collagen-I, reduce the expression of inflammatory factors in keratinocytes, and enhance skin hydration and repair effects. HA-ECT can reduce intracellular reactive oxygen species and inhibit the expression of matrix metalloproteinase-1 (reduced by 1.27-fold) to improve skin photoaging. Therefore, supramolecular HA-ECT has potential application in the field of cosmetics for skin antioxidants, anti-aging, and repair.
Collapse
Affiliation(s)
- Beibei Lu
- Department of Dermatology, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, Shenzhen 518020, Guangdong, China.
- The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen 518020, Guangdong, China
- Candidate Branch of National Clinical Research Center for Skin Diseases, Shenzhen 518020, Guangdong, China
- Department of Shenzhen People's Hospital Geriatrics Center, Shenzhen 518020, Guangdong, China
- Sauvage Laboratory for Smart Materials, Harbin Institute of Technology (Shenzhen), Shenzhen 518055, P. R. China.
- Research Centre of Printed Flexible Electronics, School of Materials Science and Engineering, Harbin Institute of Technology (Shenzhen), Shenzhen 518055, P. R. China
| | - Siran Zhao
- Division of Chemistry and Analytical Science, National Institute of Metrology, Beijing, 100029, China
| | - Jichuan Zhang
- Shenzhen Shinehigh Innovation Technology Co., Ltd., Shenzhen 518055, P. R. China
| | - Jingbo Zhan
- Shenzhen Shinehigh Innovation Technology Co., Ltd., Shenzhen 518055, P. R. China
| | - Jianglin Zhang
- Department of Dermatology, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, Shenzhen 518020, Guangdong, China.
- The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen 518020, Guangdong, China
- Candidate Branch of National Clinical Research Center for Skin Diseases, Shenzhen 518020, Guangdong, China
- Department of Shenzhen People's Hospital Geriatrics Center, Shenzhen 518020, Guangdong, China
| | - Zhe Liu
- Bloomage Biotech Co., Ltd., Jinan, Shandong 250104, China.
| | - Jiaheng Zhang
- Sauvage Laboratory for Smart Materials, Harbin Institute of Technology (Shenzhen), Shenzhen 518055, P. R. China.
- Research Centre of Printed Flexible Electronics, School of Materials Science and Engineering, Harbin Institute of Technology (Shenzhen), Shenzhen 518055, P. R. China
| |
Collapse
|
7
|
Rusiñol L, Puig L. A Narrative Review of the IL-18 and IL-37 Implications in the Pathogenesis of Atopic Dermatitis and Psoriasis: Prospective Treatment Targets. Int J Mol Sci 2024; 25:8437. [PMID: 39126010 PMCID: PMC11312859 DOI: 10.3390/ijms25158437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2024] [Revised: 07/30/2024] [Accepted: 08/01/2024] [Indexed: 08/12/2024] Open
Abstract
Atopic dermatitis and psoriasis are prevalent inflammatory skin conditions that significantly impact the quality of life of patients, with diverse treatment options available. Despite advances in understanding their underlying mechanisms, recent research highlights the significance of interleukins IL-18 and IL-37, in Th1, Th2, and Th17 inflammatory responses, closely associated with the pathogenesis of psoriasis and atopic dermatitis. Hence, IL-18 and IL-37 could potentially become therapeutic targets. This narrative review synthesizes knowledge on these interleukins, their roles in atopic dermatitis and psoriasis, and emerging treatment strategies. Findings of a literature search up to 30 May 2024, underscore a research gap in IL-37-targeted therapies. Conversely, IL-18-focused treatments have demonstrated promise in adult-onset Still's Disease, warranting further exploration for their potential efficacy in psoriasis and atopic dermatitis.
Collapse
Affiliation(s)
- Lluís Rusiñol
- Dermatology Department, Hospital de la Santa Creu i Sant Pau, 08025 Barcelona, Spain;
- Institut de Recerca Sant Pau (IR Sant Pau), Sant Quintí 77-79, 08041 Barcelona, Spain
- Unitat Docent Hospital Universitari Sant Pau, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain
| | - Lluís Puig
- Dermatology Department, Hospital de la Santa Creu i Sant Pau, 08025 Barcelona, Spain;
- Institut de Recerca Sant Pau (IR Sant Pau), Sant Quintí 77-79, 08041 Barcelona, Spain
- Unitat Docent Hospital Universitari Sant Pau, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain
| |
Collapse
|
8
|
Arrizabalaga L, Risson A, Ezcurra-Hualde M, Aranda F, Berraondo P. Unveiling the multifaceted antitumor effects of interleukin 33. Front Immunol 2024; 15:1425282. [PMID: 38881897 PMCID: PMC11176530 DOI: 10.3389/fimmu.2024.1425282] [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: 04/29/2024] [Accepted: 05/21/2024] [Indexed: 06/18/2024] Open
Abstract
Interleukin 33 (IL-33), once predominantly recognized for its pro-tumoral activities, has emerged as a multifunctional cytokine with antitumor properties. IL-33 pleiotropic activities include activation of Th1 CD4+ T cells, CD8+ T cells, NK cells, dendritic cells, eosinophils, as well as type 2 innate lymphoid cells. Regarding this immunomodulatory activity, IL-33 demonstrates synergistic interactions with various cancer therapies, including immune checkpoint blockade and chemotherapy. Combinatorial treatments leveraging IL-33 exhibit enhanced antitumor efficacy across different tumor models, promising novel avenues for cancer therapy. Despite its antitumor effects, the complex interplay of IL-33 within the tumor microenvironment underscores the need for further investigation. Understanding the mechanisms underlying IL-33's dual role as both a promoter and inhibitor of tumor progression is essential for refining therapeutic strategies and fully realizing its potential in cancer immunotherapy. This review delves into the intricate landscape of IL-33 effects within the tumor microenvironment, highlighting its pivotal role in orchestrating immune responses against cancer.
Collapse
Affiliation(s)
- Leire Arrizabalaga
- Cima Universidad de Navarra, Pamplona, Spain
- Navarra Institute for Health Research (IDISNA) and Cancer Center Clínica Universidad de Navarra (CCUN), Pamplona, Spain
| | - Aline Risson
- Cima Universidad de Navarra, Pamplona, Spain
- Navarra Institute for Health Research (IDISNA) and Cancer Center Clínica Universidad de Navarra (CCUN), Pamplona, Spain
| | - Miriam Ezcurra-Hualde
- Cima Universidad de Navarra, Pamplona, Spain
- Navarra Institute for Health Research (IDISNA) and Cancer Center Clínica Universidad de Navarra (CCUN), Pamplona, Spain
| | - Fernando Aranda
- Cima Universidad de Navarra, Pamplona, Spain
- Navarra Institute for Health Research (IDISNA) and Cancer Center Clínica Universidad de Navarra (CCUN), Pamplona, Spain
| | - Pedro Berraondo
- Cima Universidad de Navarra, Pamplona, Spain
- Navarra Institute for Health Research (IDISNA) and Cancer Center Clínica Universidad de Navarra (CCUN), Pamplona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
| |
Collapse
|
9
|
Liu D, Liu G, Liu S. Promising Application, Efficient Production, and Genetic Basis of Mannosylerythritol Lipids. Biomolecules 2024; 14:557. [PMID: 38785964 PMCID: PMC11117751 DOI: 10.3390/biom14050557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 05/01/2024] [Accepted: 05/03/2024] [Indexed: 05/25/2024] Open
Abstract
Mannosylerythritol lipids (MELs) are a class of glycolipids that have been receiving increasing attention in recent years due to their diverse biological activities. MELs are produced by certain fungi and display a range of bioactivities, making them attractive candidates for various applications in medicine, agriculture, and biotechnology. Despite their remarkable qualities, industrial-scale production of MELs remains a challenge for fungal strains. Excellent fungal strains and fermentation processes are essential for the efficient production of MELs, so efforts have been made to improve the fermentation yield by screening high-yielding strains, optimizing fermentation conditions, and improving product purification processes. The availability of the genome sequence is pivotal for elucidating the genetic basis of fungal MEL biosynthesis. This review aims to shed light on the applications of MELs and provide insights into the genetic basis for efficient MEL production. Additionally, this review offers new perspectives on optimizing MEL production, contributing to the advancement of sustainable biosurfactant technologies.
Collapse
Affiliation(s)
- Dun Liu
- College of Marine Life Science, Ocean University of China, Qingdao 266003, China;
| | - Guanglei Liu
- College of Marine Life Science, Ocean University of China, Qingdao 266003, China;
| | - Shiping Liu
- State Key Laboratory of Resource Insects, Southwest University, Beibei, Chongqing 400716, China
| |
Collapse
|
10
|
Zhao Y, Zhang J, Yang B, Li J, Ding Y, Wu L, Zhang L, Wang J, Zhu X, Zhang F, Tao X, Li Y, Zhang C, Li L, Lu J, Diao Q, Lu Q, Man X, Li F, Xia X, Cheng H, Jia Y, Zhao G, Yan J, Chen B. Efficacy and safety of CM310 in moderate-to-severe atopic dermatitis: A multicenter, randomized, double-blind, placebo-controlled phase 2b trial. Chin Med J (Engl) 2024; 137:200-208. [PMID: 37482623 PMCID: PMC10798785 DOI: 10.1097/cm9.0000000000002747] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2023] [Indexed: 07/25/2023] Open
Abstract
BACKGROUND Atopic dermatitis (AD) affects approximately 10% of adults worldwide. CM310 is a humanized monoclonal antibody targeting interleukin-4 receptor alpha that blocks interleukin-4 and interleukin-13 signaling. This trial aimed to evaluate the efficacy and safety of CM310 in Chinese adults with moderate-to-severe AD. METHODS This multicenter, randomized, double-blind, placebo-controlled, phase 2b trial was conducted in 21 medical institutions in China from February to November 2021. Totally 120 eligible patients were enrolled and randomized (1:1:1) to receive subcutaneous injections of 300 mg CM310, 150 mg CM310, or placebo every 2 weeks for 16 weeks, followed by an 8-week follow-up period. The primary endpoint was the proportion of patients achieving ≥75% improvement in the Eczema Area and Severity Index (EASI-75) score from baseline at week 16. Safety and pharmacodynamics were also studied. RESULTS At week 16, the proportion of EASI-75 responders from baseline was significantly higher in the CM310 groups (70% [28/40] for high-dose and 65% [26/40] for low-dose) than that in the placebo group (20%[8/40]). The differences in EASI-75 response rate were 50% (high vs . placebo, 95% CI 31%-69%) and 45% (low vs . placebo, 95% CI 26%-64%), with both P values <0.0001. CM310 at both doses also significantly improved the EASI score, Investigator's Global Assessment score, daily peak pruritus Numerical Rating Scale, AD-affected body surface area, and Dermatology Life Quality Index compared with placebo. CM310 treatment reduced levels of thymus and activation-regulated chemokine, total immunoglobulin E, lactate dehydrogenase, and blood eosinophils. The incidence of treatment-emergent adverse events (TEAEs) was similar among all three groups, with the most common TEAEs reported being upper respiratory tract infection, atopic dermatitis, hyperlipidemia, and hyperuricemia. No severe adverse events were deemed to be attributed to CM310. CONCLUSION CM310 at 150 mg and 300 mg every 2 weeks demonstrated significant efficacy and was well-tolerated in adults with moderate-to-severe AD.
Collapse
Affiliation(s)
- Yan Zhao
- Department of Dermatology, Peking University People's Hospital, Beijing 100044, China
| | - Jianzhong Zhang
- Department of Dermatology, Peking University People's Hospital, Beijing 100044, China
| | - Bin Yang
- Department of Dermatology, Dermatology Hospital, Southern Medical University, Guangzhou, Guangdong 510091, China
| | - Jingyi Li
- Department of Dermatology, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Yangfeng Ding
- Department of Dermatology, Shanghai Skin Disease Hospital, Institute of Psoriasis, Tongji University School of Medicine, Shanghai 200443, China
| | - Liming Wu
- Department of Dermatology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310006, China
| | - Litao Zhang
- Department of Dermatology, Tianjin Academy of Traditional Chinese Medicine Affiliated Hospital, Tianjin 300120, China
| | - Jinyan Wang
- Department of Dermatology, Ningbo No.2 Hospital, Ningbo, Zhejiang 315010, China
| | - Xiaohong Zhu
- Department of Dermatology, The Wuxi Second Affiliated Hospital of Nanjing Medical University, Wuxi, Jiangsu 214002, China
| | - Furen Zhang
- Shandong Provincial Hospital for Skin Diseases & Shandong Provincial Institute of Dermatology and Veneorology, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong 250022, China
| | - Xiaohua Tao
- Center for Plastic & Reconstructive Surgery, Department of Dermatology, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, Zhejiang 310014, China
| | - Yumei Li
- Department of Dermatology, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu 212001, China
| | - Chunlei Zhang
- Department of Dermatology, Peking University Third Hospital, Beijing 100191, China
| | - Linfeng Li
- Department of Dermatology, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Jianyun Lu
- Department of Dermatology, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, China
| | - Qingchun Diao
- Department of Dermatology, Chongqing Traditional Chinese Medicine Hospital, Chongqing 400011, China
| | - Qianjin Lu
- Hunan Key Laboratory of Medical Epigenomics, Department of Dermatology, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| | - Xiaoyong Man
- Department of Dermatology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China
| | - Fuqiu Li
- Department of Dermatology, The Second Hospital of Jilin University, Changchun, Jilin 130041, China
| | - Xiujuan Xia
- Department of Dermatology, Qingdao University Medical College Affiliated Yantai Yuhuangding Hospital, Yantai, Shandong 264000, China
| | - Hao Cheng
- Department of Dermatology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310016, China
| | - Yingmin Jia
- Clinical Department, Keymed Biosciences (Chengdu) Limited, Chengdu, Sichuan 610219, China
| | - Guoqing Zhao
- Clinical Department, Keymed Biosciences (Chengdu) Limited, Chengdu, Sichuan 610219, China
| | - Jinchun Yan
- Clinical Department, Keymed Biosciences (Chengdu) Limited, Chengdu, Sichuan 610219, China
| | - Bo Chen
- Clinical Department, Keymed Biosciences (Chengdu) Limited, Chengdu, Sichuan 610219, China
| |
Collapse
|
11
|
Yazici D, Ogulur I, Pat Y, Babayev H, Barletta E, Ardicli S, Bel Imam M, Huang M, Koch J, Li M, Maurer D, Radzikowska U, Satitsuksanoa P, Schneider SR, Sun N, Traidl S, Wallimann A, Wawrocki S, Zhakparov D, Fehr D, Ziadlou R, Mitamura Y, Brüggen MC, van de Veen W, Sokolowska M, Baerenfaller K, Nadeau K, Akdis M, Akdis CA. The epithelial barrier: The gateway to allergic, autoimmune, and metabolic diseases and chronic neuropsychiatric conditions. Semin Immunol 2023; 70:101846. [PMID: 37801907 DOI: 10.1016/j.smim.2023.101846] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Accepted: 09/27/2023] [Indexed: 10/08/2023]
Abstract
Since the 1960 s, our health has been compromised by exposure to over 350,000 newly introduced toxic substances, contributing to the current pandemic in allergic, autoimmune and metabolic diseases. The "Epithelial Barrier Theory" postulates that these diseases are exacerbated by persistent periepithelial inflammation (epithelitis) triggered by exposure to a wide range of epithelial barrier-damaging substances as well as genetic susceptibility. The epithelial barrier serves as the body's primary physical, chemical, and immunological barrier against external stimuli. A leaky epithelial barrier facilitates the translocation of the microbiome from the surface of the afflicted tissues to interepithelial and even deeper subepithelial locations. In turn, opportunistic bacterial colonization, microbiota dysbiosis, local inflammation and impaired tissue regeneration and remodelling follow. Migration of inflammatory cells to susceptible tissues contributes to damage and inflammation, initiating and aggravating many chronic inflammatory diseases. The objective of this review is to highlight and evaluate recent studies on epithelial physiology and its role in the pathogenesis of chronic diseases in light of the epithelial barrier theory.
Collapse
Affiliation(s)
- Duygu Yazici
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - 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
| | - Huseyn Babayev
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Elena Barletta
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland; Swiss Institute of Bioinformatics (SIB), Davos, Switzerland
| | - Sena Ardicli
- 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
| | - Mengting Huang
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Jana Koch
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland; Swiss Institute of Bioinformatics (SIB), Davos, Switzerland
| | - Manru Li
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Debbie Maurer
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Urszula Radzikowska
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland; Christine Kühne-Center for Allergy Research and Education, Davos, Switzerland
| | | | - Stephan R Schneider
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Na Sun
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland; National Engineering Research Center of Seafood, School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, PR China
| | - Stephan Traidl
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland; Division of Immunodermatology and Allergy Research, Department of Dermatology and Allergy, Hannover Medical School, Hannover, Germany
| | - Alexandra Wallimann
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Sebastian Wawrocki
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Damir Zhakparov
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Danielle Fehr
- Christine Kühne-Center for Allergy Research and Education, Davos, Switzerland; Faculty of Medicine, University of Zurich, Zurich, Switzerland; Department of Dermatology, University Hospital Zurich, Zurich, Switzerland
| | - Reihane Ziadlou
- Christine Kühne-Center for Allergy Research and Education, Davos, Switzerland; Faculty of Medicine, University of Zurich, Zurich, Switzerland; Department of Dermatology, University Hospital Zurich, Zurich, Switzerland
| | - Yasutaka Mitamura
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Marie-Charlotte Brüggen
- Christine Kühne-Center for Allergy Research and Education, Davos, Switzerland; Faculty of Medicine, University of Zurich, Zurich, Switzerland; Department of Dermatology, University Hospital Zurich, Zurich, Switzerland
| | - Willem van de Veen
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland; Christine Kühne-Center for Allergy Research and Education, Davos, Switzerland
| | - Milena Sokolowska
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland; Christine Kühne-Center for Allergy Research and Education, Davos, Switzerland
| | - Katja Baerenfaller
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland; Swiss Institute of Bioinformatics (SIB), Davos, Switzerland
| | - Kari Nadeau
- Department of Environmental Health, T.H. Chan School of Public Health, Harvard University, 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; Christine Kühne-Center for Allergy Research and Education, Davos, Switzerland.
| |
Collapse
|
12
|
Wang Z, Tang N. Unpacking the complexity of nuclear IL-33 (nIL-33): a crucial regulator of transcription and signal transduction. J Cell Commun Signal 2023:10.1007/s12079-023-00788-1. [PMID: 37878185 DOI: 10.1007/s12079-023-00788-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Accepted: 10/12/2023] [Indexed: 10/26/2023] Open
Abstract
Interleukin-33 (IL-33) (NF-HEV), a chromatin-associated nuclear cytokine, is a member of the IL-1 family. IL-33 possesses a nuclear localization signal and a homeodomain (a structure resembling a helix-turn-helix) that can bind to nuclear chromatin. Research has revealed that IL-33 can function as a nuclear factor to regulate various biological processes. This review discusses the cellular localization, functional effects, and immune regulation of full length IL-33 (FLIL-33), cytokine IL-33 (sIL-33) and nuclear IL-33 (nIL-33). In addition, the post-translational modifications of nIL-33 and the hypothesis of using nIL-33 as a treatment method were also summarized. A multidisciplinary approach is required which integrates methods and techniques from genomics, proteomics, cell biology and immunology to provide comprehensive insights into the function and therapeutic potential of nIL-33.
Collapse
Affiliation(s)
- Zengbin Wang
- Department of Immunology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Nanhong Tang
- Department of Hepatobiliary Surgery, Fujian Institute of Hepatobiliary Surgery, Fujian Medical University Union Hospital, Fuzhou, China.
| |
Collapse
|
13
|
Tsuji G, Yamamura K, Kawamura K, Kido-Nakahara M, Ito T, Nakahara T. Regulatory Mechanism of the IL-33-IL-37 Axis via Aryl Hydrocarbon Receptor in Atopic Dermatitis and Psoriasis. Int J Mol Sci 2023; 24:14633. [PMID: 37834081 PMCID: PMC10572928 DOI: 10.3390/ijms241914633] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 09/20/2023] [Accepted: 09/26/2023] [Indexed: 10/15/2023] Open
Abstract
Interleukin (IL)-33 and IL-37 have been identified as novel cytokines involved in various inflammatory diseases. However, their specific roles remain largely unknown. Recent studies have shown that IL-33, which triggers inflammation, and IL-37, which suppresses it, cooperatively regulate the balance between inflammation and anti-inflammation. IL-33 and IL-37 are also deeply involved in the pathogenesis of inflammatory skin diseases such as atopic dermatitis (AD) and psoriasis. Furthermore, a signaling pathway by which aryl hydrocarbon receptor (AHR), a receptor for dioxins, regulates the expression of IL-33 and IL-37 has been revealed. Here, we outline recent findings on the mechanisms regulating IL-33 and IL-37 expression in AD and psoriasis. IL-33 expression is partially dependent on mitogen-activated protein kinase (MAPK) activation, and IL-37 has a role in suppressing MAPK in human keratinocytes. Furthermore, IL-33 downregulates skin barrier function proteins including filaggrin and loricrin, thereby downregulating the expression of IL-37, which colocalizes with these proteins. This leads to an imbalance of the IL-33-IL-37 axis, involving increased IL-33 and decreased IL-37, which may be associated with the pathogenesis of AD and psoriasis. Therefore, AHR-mediated regulation of the IL-33-IL-37 axis may lead to new therapeutic strategies for the treatment of AD and psoriasis.
Collapse
Affiliation(s)
- Gaku Tsuji
- Research and Clinical Center for Yusho and Dioxin, Kyushu University Hospital, 3-1-1, Maidashi, Higashi-ku, Fukuoka 812-8582, Japan; (K.Y.); (T.N.)
- Department of Dermatology, Graduate School of Medical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-ku, Fukuoka 812-8582, Japan; (K.K.); (M.K.-N.); (T.I.)
| | - Kazuhiko Yamamura
- Research and Clinical Center for Yusho and Dioxin, Kyushu University Hospital, 3-1-1, Maidashi, Higashi-ku, Fukuoka 812-8582, Japan; (K.Y.); (T.N.)
- Department of Dermatology, Graduate School of Medical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-ku, Fukuoka 812-8582, Japan; (K.K.); (M.K.-N.); (T.I.)
| | - Koji Kawamura
- Department of Dermatology, Graduate School of Medical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-ku, Fukuoka 812-8582, Japan; (K.K.); (M.K.-N.); (T.I.)
| | - Makiko Kido-Nakahara
- Department of Dermatology, Graduate School of Medical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-ku, Fukuoka 812-8582, Japan; (K.K.); (M.K.-N.); (T.I.)
| | - Takamichi Ito
- Department of Dermatology, Graduate School of Medical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-ku, Fukuoka 812-8582, Japan; (K.K.); (M.K.-N.); (T.I.)
| | - Takeshi Nakahara
- Research and Clinical Center for Yusho and Dioxin, Kyushu University Hospital, 3-1-1, Maidashi, Higashi-ku, Fukuoka 812-8582, Japan; (K.Y.); (T.N.)
- Department of Dermatology, Graduate School of Medical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-ku, Fukuoka 812-8582, Japan; (K.K.); (M.K.-N.); (T.I.)
| |
Collapse
|
14
|
Hatano Y, Elias PM. "Outside-to-inside," "inside-to-outside," and "intrinsic" endogenous pathogenic mechanisms in atopic dermatitis: keratinocytes as the key functional cells involved in both permeability barrier dysfunction and immunological alterations. Front Immunol 2023; 14:1239251. [PMID: 37638035 PMCID: PMC10451066 DOI: 10.3389/fimmu.2023.1239251] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Accepted: 07/31/2023] [Indexed: 08/29/2023] Open
Abstract
Permeability barrier disruption has been shown to induce immunological alterations (i.e., an "outside-to-inside" pathogenic mechanism). Conversely, several inflammatory and immunological mechanisms reportedly interrupt permeability barrier homeostasis (i.e., an "inside-to-outside" pathogenic mechanism). It is now widely recognized that alterations of even a single molecule in keratinocytes can lead to not only permeability barrier dysfunction but also to immunological alterations. Such a simultaneous, bidirectional functional change by keratinocytes is herein named an "intrinsic" pathogenic mechanism. Molecules and/or pathways involved in this mechanism could be important not only as factors in disease pathogenesis but also as potential therapeutic targets for inflammatory cutaneous diseases, such as atopic dermatitis, psoriasis, and prurigo nodularis. Elevation of skin surface pH following permeability barrier abrogation comprises one of the key pathogenic phenomena of the "outside-to-inside" mechanism. Not only type 2 cytokines (e.g., IL-4, IL-13, IL-31) but also type 1 (e.g. IFN-γ), and type 3 (e.g., IL-17, IL-22) as well as several other inflammatory factors (e.g. histamine) can disrupt permeability barrier homeostasis and are all considered part of the "inside-to-outside" mechanism. Finally, examples of molecules relevant to the "intrinsic" pathogenic mechanism include keratin 1, filaggrin, and peroxisome proliferator-activated receptor-α (PPARα).
Collapse
Affiliation(s)
- Yutaka Hatano
- Department of Dermatology, Faculty of Medicine, Oita University, Oita, Japan
| | - Peter M. Elias
- Department of Dermatology, University of California, San Francisco and Veterans Affairs Health Care System, San Francisco, CA, United States
| |
Collapse
|
15
|
Dai X, Shiraishi K, Muto J, Mori H, Murakami M, Sayama K. Nuclear IL-33 Plays an Important Role in EGFR-Mediated Keratinocyte Migration by Regulating the Activation of Signal Transducer and Activator of Transcription 3 and NF-κB. JID INNOVATIONS 2023; 3:100205. [PMID: 37441125 PMCID: PMC10333683 DOI: 10.1016/j.xjidi.2023.100205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 02/21/2023] [Accepted: 02/27/2023] [Indexed: 07/15/2023] Open
Abstract
Nuclear IL-33 levels are high at the epidermal edges of skin wounds and facilitate wound healing. However, IL-33-mediated regulation of keratinocyte (KC) biology during wound healing remains poorly understood. During skin-wound healing, KC migration and re-epithelialization are mediated predominantly by EGFR signaling activation and depend on the function of signal transducer and activator of transcription 3 (STAT3). We found that migrating KCs at the leading edges of mouse skin wounds exhibited concomitant induction and nuclear colocalization of IL-33 and phosphorylated STAT3. In cultured human KCs, activation of EGFR signaling caused rapid elevation of nuclear IL-33, which directly interacts with phosphorylated STAT3, promoting STAT3 activation. In vitro KC migration and wound-healing assays revealed that high nuclear IL-33 levels were required for KC migration and wound closure. KC mobility associated with a lack of suprabasal epidermal keratins and extracellular matrix degradation mediated by matrix metalloproteinases (MMPs) control cell migration at the intracellular and extracellular levels, respectively. In EGFR-activated KCs, nuclear IL-33 mediated keratin 1 and 10 downregulation and MMP9 upregulation by promoting STAT3 activation and limited MMP1, MMP3, and MMP10 induction by suppressing NF-κB transactivation. Thus, epidermal nuclear IL-33 is involved in KC migration and wound closure by regulating the STAT3 and NF-κB pathways.
Collapse
Affiliation(s)
- Xiuju Dai
- Department of Dermatology, Ehime University Graduate School of Medicine, Ehime, Japan
| | - Ken Shiraishi
- Department of Dermatology, Ehime University Graduate School of Medicine, Ehime, Japan
| | - Jun Muto
- Department of Dermatology, Ehime University Graduate School of Medicine, Ehime, Japan
| | - Hideki Mori
- Department of Dermatology, Ehime University Graduate School of Medicine, Ehime, Japan
| | - Masamoto Murakami
- Department of Dermatology, Ehime University Graduate School of Medicine, Ehime, Japan
| | - Koji Sayama
- Department of Dermatology, Ehime University Graduate School of Medicine, Ehime, Japan
| |
Collapse
|
16
|
A Kaleidoscope of Keratin Gene Expression and the Mosaic of Its Regulatory Mechanisms. Int J Mol Sci 2023; 24:ijms24065603. [PMID: 36982676 PMCID: PMC10052683 DOI: 10.3390/ijms24065603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 03/07/2023] [Accepted: 03/10/2023] [Indexed: 03/17/2023] Open
Abstract
Keratins are a family of intermediate filament-forming proteins highly specific to epithelial cells. A combination of expressed keratin genes is a defining property of the epithelium belonging to a certain type, organ/tissue, cell differentiation potential, and at normal or pathological conditions. In a variety of processes such as differentiation and maturation, as well as during acute or chronic injury and malignant transformation, keratin expression undergoes switching: an initial keratin profile changes accordingly to changed cell functions and location within a tissue as well as other parameters of cellular phenotype and physiology. Tight control of keratin expression implies the presence of complex regulatory landscapes within the keratin gene loci. Here, we highlight patterns of keratin expression in different biological conditions and summarize disparate data on mechanisms controlling keratin expression at the level of genomic regulatory elements, transcription factors (TFs), and chromatin spatial structure.
Collapse
|
17
|
Safe and Effective Antioxidant: The Biological Mechanism and Potential Pathways of Ergothioneine in the Skin. Molecules 2023; 28:molecules28041648. [PMID: 36838636 PMCID: PMC9967237 DOI: 10.3390/molecules28041648] [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: 01/06/2023] [Revised: 02/02/2023] [Accepted: 02/04/2023] [Indexed: 02/11/2023] Open
Abstract
Ergothioneine, a sulfur-containing micromolecular histidine derivative, has attracted increasing attention from scholars since it was confirmed in the human body. In the human body, ergothioneine is transported and accumulated specifically through OCTN-1, especially in the mitochondria and nucleus, suggesting that it can target damaged cells and tissues as an antioxidant. It shows excellent antioxidant, anti-inflammatory effects, and anti-aging properties, and inhibits melanin production. It is a mega antioxidant that may participate in the antioxidant network system and promote the reducing glutathione regeneration cycle. This review summarizes studies on the antioxidant effects of ergothioneine on various free radicals in vitro to date and systematically introduces its biological activities and potential mechanisms, mostly in dermatology. Additionally, the application of ergothioneine in cosmetics is briefly summarized. Lastly, we propose some problems that require solutions to understand the mechanism of action of ergothioneine. We believe that ergothioneine has good prospects in the food and cosmetics industries, and can thus meet some needs of the health and beauty industry.
Collapse
|
18
|
Luzina IG, Lockatell V, Courneya JP, Mei Z, Fishelevich R, Kopach P, Pickering EM, Kang PH, Krupnick AS, Todd NW, Vogel SN, Atamas SP. Full-length IL-33 augments pulmonary fibrosis in an ST2- and Th2-independent, non-transcriptomic fashion. Cell Immunol 2023; 383:104657. [PMID: 36603504 PMCID: PMC9909894 DOI: 10.1016/j.cellimm.2022.104657] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 11/07/2022] [Accepted: 12/13/2022] [Indexed: 12/23/2022]
Abstract
Mature IL-33 (MIL33) acting through its receptor, ST2, is known to regulate fibrosis. The precursor, full-length IL-33 (FLIL33), may function differently from MIL33 and independently of ST2. Here we report that genetic deletion of either IL-33 or ST2 attenuates pulmonary fibrosis in the bleomycin model, as does Cre-induced IL-33 deficiency in response to either acute or chronic bleomycin challenge. However, adenovirus-mediated gene delivery of FLIL33, but not MIL33, to the lungs of either wild-type or ST2-deficient mice potentiates the profibrotic effect of bleomycin without inducing a Th2 phenotype. In cultured mouse lung cells, FLIL33 overexpression induces moderate and distinct transcriptomic changes compared with a robust response induced by MIL33, whereas ST2 deletion abrogates the effects of both IL-33 forms. Thus, FLIL33 may contribute to fibrosis in an ST2-independent, Th2-independent, non-transcriptomic fashion, suggesting that pharmacological targeting of both FLIL33 and MIL33 may prove efficacious in patients with pulmonary fibrosis.
Collapse
Affiliation(s)
- Irina G Luzina
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, United States; Research Service, Baltimore VA Medical Center, Baltimore, MD, United States.
| | - Virginia Lockatell
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Jean-Paul Courneya
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Zhongcheng Mei
- Department of Surgery, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Rita Fishelevich
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Pavel Kopach
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Edward M Pickering
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Phillip H Kang
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Alexander S Krupnick
- Research Service, Baltimore VA Medical Center, Baltimore, MD, United States; Department of Surgery, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Nevins W Todd
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, United States; Research Service, Baltimore VA Medical Center, Baltimore, MD, United States
| | - Stefanie N Vogel
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Sergei P Atamas
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, United States; Research Service, Baltimore VA Medical Center, Baltimore, MD, United States; Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, United States
| |
Collapse
|
19
|
Mießner H, Seidel J, Smith ESJ. In vitro models for investigating itch. Front Mol Neurosci 2022; 15:984126. [PMID: 36385768 PMCID: PMC9644192 DOI: 10.3389/fnmol.2022.984126] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 10/10/2022] [Indexed: 12/04/2022] Open
Abstract
Itch (pruritus) is a sensation that drives a desire to scratch, a behavior observed in many animals. Although generally short-lasting and not causing harm, there are several pathological conditions where chronic itch is a hallmark symptom and in which prolonged scratching can induce damage. Finding medications to counteract the sensation of chronic itch has proven difficult due to the molecular complexity that involves a multitude of triggers, receptors and signaling pathways between skin, immune and nerve cells. While much has been learned about pruritus from in vivo animal models, they have limitations that corroborate the necessity for a transition to more human disease-like models. Also, reducing animal use should be encouraged in research. However, conducting human in vivo experiments can also be ethically challenging. Thus, there is a clear need for surrogate models to be used in pre-clinical investigation of the mechanisms of itch. Most in vitro models used for itch research focus on the use of known pruritogens. For this, sensory neurons and different types of skin and/or immune cells are stimulated in 2D or 3D co-culture, and factors such as neurotransmitter or cytokine release can be measured. There are however limitations of such simplistic in vitro models. For example, not all naturally occurring cell types are present and there is also no connection to the itch-sensing organ, the central nervous system (CNS). Nevertheless, in vitro models offer a chance to investigate otherwise inaccessible specific cell–cell interactions and molecular pathways. In recent years, stem cell-based approaches and human primary cells have emerged as viable alternatives to standard cell lines or animal tissue. As in vitro models have increased in their complexity, further opportunities for more elaborated means of investigating itch have been developed. In this review, we introduce the latest concepts of itch and discuss the advantages and limitations of current in vitro models, which provide valuable contributions to pruritus research and might help to meet the unmet clinical need for more refined anti-pruritic substances.
Collapse
Affiliation(s)
- Hendrik Mießner
- Department of Pharmacology, University of Cambridge, Cambridge, United Kingdom
- Dermatological Skin Care, Beiersdorf AG, Hamburg, Germany
| | - Judith Seidel
- Dermatological Skin Care, Beiersdorf AG, Hamburg, Germany
| | - Ewan St. John Smith
- Department of Pharmacology, University of Cambridge, Cambridge, United Kingdom
- *Correspondence: Ewan St. John Smith,
| |
Collapse
|
20
|
Lv WJ, Huang JY, Li SP, Gong XP, Sun JB, Mao W, Guo SN. Portulaca oleracea L. extracts alleviate 2,4-dinitrochlorobenzene-induced atopic dermatitis in mice. Front Nutr 2022; 9:986943. [PMID: 36051905 PMCID: PMC9424637 DOI: 10.3389/fnut.2022.986943] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Accepted: 07/28/2022] [Indexed: 11/18/2022] Open
Abstract
Atopic dermatitis (AD) is a common chronic allergic skin disease characterized clinically by severe skin lesions and pruritus. Portulaca oleracea L. (PO) is a resourceful plant with homologous properties in medicine and food. In this study, we used two different methods to extract PO, and compared the therapeutic effects of PO aqueous extract (POAE) and PO ultrasound-assisted ethanol extract (POEE) on 2,4-dinitrochlorobenzene (DNCB)-induced AD mice. The results showed that in POAE and POEE, the extraction rates of polysaccharides were 16.95% and 9.85%, while the extraction rates of total flavonoids were 3.15% and 3.25%, respectively. Compared with AD mice, clinical symptoms such as erythema, edema, dryness and ulceration in the back and left ear were alleviated, and pruritus behavior was reduced after POAE and POEE treatments. The thickness of the skin epidermis was thinned, the density of skin nerve fibers labeled with protein gene product 9.5 (PGP9.5) was decreased, and mast cell infiltration was reduced. There was a decrease in blood lymphocytes, eosinophils and basophils, a significant decrease in spleen index and a noticeable decrease in serum immunoglobulin E (Ig E). POEE significantly reduced the concentration of the skin pruritic factor interleukin (Il)-31. POAE and POEE reduced the concentration of skin histamine (His), down-regulated mRNA expression levels of interferon-γ (Ifnγ), tumor necrosis factor-α (Tnf-α), thymic stromal lymphopoietin (Tslp) and Il-4, with an increase of Filaggrin (Flg) and Loricrin (Lor) in skin lesions. These results suggested that POAE and POEE may inhibit atopic response and alleviate the clinical symptoms of AD by inhibiting the expression of immune cells, inflammatory mediators and cytokines. PO may be a potential effective drug for AD-like diseases.
Collapse
Affiliation(s)
- Wei-jie Lv
- College of Veterinary Medicine, South China Agricultural University, State Key Laboratory of Dampness Syndrome of Chinese Medicine, Guangzhou, China
| | - Jie-yi Huang
- College of Veterinary Medicine, South China Agricultural University, State Key Laboratory of Dampness Syndrome of Chinese Medicine, Guangzhou, China
| | - Shu-peng Li
- College of Veterinary Medicine, South China Agricultural University, State Key Laboratory of Dampness Syndrome of Chinese Medicine, Guangzhou, China
| | - Xiao-pei Gong
- College of Veterinary Medicine, South China Agricultural University, State Key Laboratory of Dampness Syndrome of Chinese Medicine, Guangzhou, China
| | - Jing-bo Sun
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, South China Agricultural University, Guangzhou, China
- *Correspondence: Jing-bo Sun,
| | - Wei Mao
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, South China Agricultural University, Guangzhou, China
- Wei Mao,
| | - Shi-ning Guo
- College of Veterinary Medicine, South China Agricultural University, State Key Laboratory of Dampness Syndrome of Chinese Medicine, Guangzhou, China
- Guangdong Technology Research Center for Traditional Chinese Veterinary Medicine and Natural Medicine, Guangzhou, China
- Guangdong Provincial Key Laboratory of Prevention and Control for Severe Clinical Animal Diseases, Guangzhou, China
- International Institute of Traditional Chinese Veterinary Medicine, Guangzhou, China
- Shi-ning Guo,
| |
Collapse
|
21
|
Jing C, Guo J, Li Z, Xu X, Wang J, Zhai L, Liu J, Sun G, Wang F, Xu Y, Li Z, Zhao D, Jiang R, Sun L. Screening and Research on Skin Barrier Damage Protective Efficacy of Different Mannosylerythritol Lipids. Molecules 2022; 27:molecules27144648. [PMID: 35889520 PMCID: PMC9320248 DOI: 10.3390/molecules27144648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2022] [Revised: 07/09/2022] [Accepted: 07/19/2022] [Indexed: 02/05/2023] Open
Abstract
Mannosylerythritol lipids (MELs) may prevent skin barrier damage, although their protective mechanisms and active monomeric constituents remain unclear. Here, three MELs were extracted from Candida antarctica cultures containing fermented olive oil then purified using silica gel-based column chromatography and semipreparative HPLC. All three compounds (MEL-A, MEL-B, MEL-C) were well separated and stable, and reliable materials were used for NMR and HRESIMS chemical structure determinations and for assessing MELs’ protective effects against skin damage. Notably, MEL-B and MEL-C effectively protected HaCaT cells from UVB-induced damage by upregulating the contents of filaggrin (FLG) and transglutaminase-1 (TGM1), as determined via ELISA. Moreover, MEL-B treatment (20 μg/mL) of UVB-irradiated HaCaT cells led to the upregulation of both the expression of mRNA genes and the key proteins FLG, LOR, and TGM1, which are known to be decreased in damaged skin cells. Additionally, histopathological analysis results revealed a markedly reduced intracellular vacuolation and cell damage, reflecting improved skin function after MEL-B treatment. Furthermore, immunofluorescence results revealed that MEL-B protected EpiKutis® three-dimensional cultured human skin cells from sodium dodecyl sulfate-induced damage by up-regulating FLG, LOR, and TGM1 expression. Accordingly, MELs’ protection against skin barrier damage depended on MEL-B monomeric constituent activities, thus highlighting their promise as beneficial ingredients for use in skin-care products.
Collapse
Affiliation(s)
- Chenxu Jing
- Research Center of Traditional Chinese Medicine, The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun 130021, China; (C.J.); (Z.L.); (X.X.); (J.W.); (L.Z.); (G.S.); (F.W.)
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun 130117, China; (J.G.); (J.L.); (D.Z.)
| | - Jiling Guo
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun 130117, China; (J.G.); (J.L.); (D.Z.)
| | - Zhenzhuo Li
- Research Center of Traditional Chinese Medicine, The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun 130021, China; (C.J.); (Z.L.); (X.X.); (J.W.); (L.Z.); (G.S.); (F.W.)
| | - Xiaohao Xu
- Research Center of Traditional Chinese Medicine, The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun 130021, China; (C.J.); (Z.L.); (X.X.); (J.W.); (L.Z.); (G.S.); (F.W.)
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun 130117, China; (J.G.); (J.L.); (D.Z.)
| | - Jing Wang
- Research Center of Traditional Chinese Medicine, The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun 130021, China; (C.J.); (Z.L.); (X.X.); (J.W.); (L.Z.); (G.S.); (F.W.)
| | - Lu Zhai
- Research Center of Traditional Chinese Medicine, The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun 130021, China; (C.J.); (Z.L.); (X.X.); (J.W.); (L.Z.); (G.S.); (F.W.)
| | - Jianzeng Liu
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun 130117, China; (J.G.); (J.L.); (D.Z.)
| | - Guang Sun
- Research Center of Traditional Chinese Medicine, The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun 130021, China; (C.J.); (Z.L.); (X.X.); (J.W.); (L.Z.); (G.S.); (F.W.)
| | - Fei Wang
- Research Center of Traditional Chinese Medicine, The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun 130021, China; (C.J.); (Z.L.); (X.X.); (J.W.); (L.Z.); (G.S.); (F.W.)
| | - Yangfen Xu
- Modern Hanfang Technology Company Limited, Guangzhou 510550, China; (Y.X.); (Z.L.)
| | - Zhaolian Li
- Modern Hanfang Technology Company Limited, Guangzhou 510550, China; (Y.X.); (Z.L.)
| | - Daqing Zhao
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun 130117, China; (J.G.); (J.L.); (D.Z.)
| | - Rui Jiang
- Research Center of Traditional Chinese Medicine, The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun 130021, China; (C.J.); (Z.L.); (X.X.); (J.W.); (L.Z.); (G.S.); (F.W.)
- Correspondence: (R.J.); (L.S.)
| | - Liwei Sun
- Research Center of Traditional Chinese Medicine, The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun 130021, China; (C.J.); (Z.L.); (X.X.); (J.W.); (L.Z.); (G.S.); (F.W.)
- Correspondence: (R.J.); (L.S.)
| |
Collapse
|
22
|
Yeoh WJ, Vu VP, Krebs P. IL-33 biology in cancer: An update and future perspectives. Cytokine 2022; 157:155961. [PMID: 35843125 DOI: 10.1016/j.cyto.2022.155961] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 05/03/2022] [Accepted: 07/01/2022] [Indexed: 12/14/2022]
Abstract
Interleukin-33 (IL-33) is a member of the IL-1 family of cytokines that is constitutively expressed in the nucleus of epithelial, endothelial and fibroblast-like cells. Upon cell stress, damage or necrosis, IL-33 is released into the cytoplasm to exert its prime role as an alarmin by binding to its specific receptor moiety, ST2. IL-33 exhibits pleiotropic function in inflammatory diseases and particularly in cancer. IL-33 may play a dual role as both a pro-tumorigenic and anti-tumorigenic cytokine, dependent on tumor and cellular context, expression levels, bioactivity and the nature of the inflammatory environment. In this review, we discuss the differential contribution of IL-33 to malignant or inflammatory conditions, its multifaceted effects on the tumor microenvironment, while providing possible explanations for the discrepant findings described in the literature. Additionally, we examine the emerging and divergent functions of IL-33 in the nucleus, and aspects of IL-33 biology that are currently under-addressed.
Collapse
Affiliation(s)
- Wen Jie Yeoh
- Institute of Pathology, University of Bern, Bern, Switzerland; Graduate School for Cellular and Biomedical Sciences, University of Bern, Switzerland
| | - Vivian P Vu
- Institute of Pathology, University of Bern, Bern, Switzerland; Graduate School for Cellular and Biomedical Sciences, University of Bern, Switzerland
| | - Philippe Krebs
- Institute of Pathology, University of Bern, Bern, Switzerland.
| |
Collapse
|
23
|
Beck LA, Cork MJ, Amagai M, De Benedetto A, Kabashima K, Hamilton JD, Rossi AB. Type 2 Inflammation Contributes to Skin Barrier Dysfunction in Atopic Dermatitis. JID INNOVATIONS 2022; 2:100131. [PMID: 36059592 PMCID: PMC9428921 DOI: 10.1016/j.xjidi.2022.100131] [Citation(s) in RCA: 108] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 01/04/2022] [Accepted: 01/06/2022] [Indexed: 01/02/2023] Open
Abstract
Skin barrier dysfunction, a defining feature of atopic dermatitis (AD), arises from multiple interacting systems. In AD, skin inflammation is caused by host-environment interactions involving keratinocytes as well as tissue-resident immune cells such as type 2 innate lymphoid cells, basophils, mast cells, and T helper type 2 cells, which produce type 2 cytokines, including IL-4, IL-5, IL-13, and IL-31. Type 2 inflammation broadly impacts the expression of genes relevant for barrier function, such as intracellular structural proteins, extracellular lipids, and junctional proteins, and enhances Staphylococcus aureus skin colonization. Systemic anti‒type 2 inflammation therapies may improve dysfunctional skin barrier in AD.
Collapse
Key Words
- AD, atopic dermatitis
- AMP, antimicrobial peptide
- CLDN, claudin
- FFA, free fatty acid
- ILC2, type 2 innate lymphoid cell
- Jaki, Jak inhibitor
- K, keratin
- KC, keratinocyte
- MMP, matrix metalloproteinase
- NMF, natural moisturizing factor
- PAR, protease-activated receptor
- PDE-4, phosphodiesterase-4
- SC, stratum corneum
- SG, stratum granulosum
- TCI, topical calcineurin inhibitor
- TCS, topical corticosteroid
- TEWL, transepidermal water loss
- TJ, tight junction
- TLR, toll-like receptor
- TNF-α, tumor necrosis factor alpha
- TYK, tyrosine kinase
- Th, T helper
- ZO, zona occludens
- hBD, human β-defensin
Collapse
Affiliation(s)
- Lisa A. Beck
- Department of Dermatology, University of Rochester Medical Center, Rochester, New York, USA,Correspondence: Lisa A. Beck, Department of Dermatology, University of Rochester Medical Center, 601 Elmwood Ave, Box 697, Rochester, New York 14642, USA.
| | - Michael J. Cork
- Sheffield Dermatology Research, Department of Infection, Immunity and Cardiovascular Disease (IICD), The University of Sheffield, The Medical School, Sheffield, United Kingdom
| | - Masayuki Amagai
- Department of Dermatology, Keio University School of Medicine, Tokyo, Japan,Laboratory for Skin Homeostasis, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Anna De Benedetto
- Department of Dermatology, University of Rochester Medical Center, Rochester, New York, USA
| | - Kenji Kabashima
- Department of Dermatology, Kyoto University Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | | | | |
Collapse
|
24
|
Dai X, Murakami M, Shiraishi K, Muto J, Tohyama M, Mori H, Utsunomiya R, Sayama K. EGFR ligands synergistically increase IL-17A-induced expression of psoriasis signature genes in human keratinocytes via IκBζ and Bcl3. Eur J Immunol 2022; 52:994-1005. [PMID: 35411943 DOI: 10.1002/eji.202149706] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Revised: 04/11/2022] [Accepted: 04/11/2022] [Indexed: 11/07/2022]
Abstract
Various epidermal growth factor receptor (EGFR) ligands are highly expressed in the epidermis of psoriasis lesions, and abnormal EGFR activation appears to be involved in the pathogenesis of psoriasis. However, how EGFR signaling contributes to the development of psoriasis is unclear. Interleukin (IL)-17A, a critical effector of the IL-23/IL-17A pathway, increases the expression of psoriasis signature genes in keratinocytes and plays an essential role in the pathogenesis of psoriasis by inducing IκBζ, a critical transcriptional regulator in psoriasis. In this study, we stimulated primary human keratinocytes with IL-17A and various EGFR ligands to investigate whether EGFR ligands regulate the expression of psoriasis signature genes. In cultured normal human keratinocytes and a living skin equivalent, EGFR ligands did not induce psoriasis-related gene expression, but significantly enhanced the IL-17A-mediated induction of various psoriasis signature genes, including antimicrobial peptides, cytokines, and chemokines. This was dependent on an EGFR activation-mediated synergistic increase in IL-17A-induced IκBζ expression and was partially mediated by the EGFR-dependent upregulation of Bcl3. Therefore, EGFR ligands can act as synergistic agents of IL-17A signaling by stimulating the epidermal production of psoriasis signature genes in psoriasis lesions. This study reveals a potential mechanism by which EGFR signaling contributes to the pathogenesis of psoriasis. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Xiuju Dai
- Department of Dermatology, Ehime University Graduate School of Medicine, Ehime, Japan
| | - Masamoto Murakami
- Department of Dermatology, Ehime University Graduate School of Medicine, Ehime, Japan
| | - Ken Shiraishi
- Department of Dermatology, Ehime University Graduate School of Medicine, Ehime, Japan
| | - Jun Muto
- Department of Dermatology, Ehime University Graduate School of Medicine, Ehime, Japan
| | - Mikiko Tohyama
- Department of Dermatology, Ehime University Graduate School of Medicine, Ehime, Japan.,Department of Dermatology, National Hospital Organization Shikoku Cancer Center, Matsuyama, Ehime, Japan
| | - Hideki Mori
- Department of Dermatology, Ehime University Graduate School of Medicine, Ehime, Japan
| | - Ryo Utsunomiya
- Department of Dermatology, Ehime University Graduate School of Medicine, Ehime, Japan
| | - Koji Sayama
- Department of Dermatology, Ehime University Graduate School of Medicine, Ehime, Japan
| |
Collapse
|
25
|
Dai X, Muto J, Shiraishi K, Utsunomiya R, Mori H, Murakami M, Sayama K. TSLP impairs epidermal barrier integrity by stimulating the formation of nuclear IL-33/phosphorylated STAT3 complex in human keratinocytes. J Invest Dermatol 2022; 142:2100-2108.e5. [DOI: 10.1016/j.jid.2022.01.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 12/20/2021] [Accepted: 01/04/2022] [Indexed: 12/20/2022]
|
26
|
Mestrallet G, Rouas-Freiss N, LeMaoult J, Fortunel NO, Martin MT. Skin Immunity and Tolerance: Focus on Epidermal Keratinocytes Expressing HLA-G. Front Immunol 2021; 12:772516. [PMID: 34938293 PMCID: PMC8685247 DOI: 10.3389/fimmu.2021.772516] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Accepted: 11/16/2021] [Indexed: 12/27/2022] Open
Abstract
Although the role of epidermal cells in skin regeneration has been extensively documented, their functions in immunity and tolerance mechanisms are largely underestimated. The aim of the present review was to outline the state of knowledge on resident immune cells of hematopoietic origin hosted in the epidermis, and then to focus on the involvement of keratinocytes in the complex skin immune networks acting in homeostasis and regeneration conditions. Based on this knowledge, the mechanisms of immune tolerance are reviewed. In particular, strategies based on immunosuppression mediated by HLA-G are highlighted, as recent advances in this field open up perspectives in epidermis-substitute bioengineering for temporary and permanent skin replacement strategies.
Collapse
Affiliation(s)
- Guillaume Mestrallet
- Commissariat ã l'Energie Atomique et aux Energies Alternatives, DRF, Francois Jacob Institute of Biology, Laboratory of Genomics and Radiobiology of Keratinopoiesis, Institute of Cellular and Molecular Radiobiology, Evry, France.,Université Paris-Saclay, Saint-Aubin, France
| | - Nathalie Rouas-Freiss
- Commissariat ã l'Energie Atomique et aux Energies Alternatives, DRF, Francois Jacob Institute of Biology, Hemato-Immunology Research Department, Saint-Louis Hospital, Paris, France.,Université de Paris, UMR-S 976 HIPI Unit, Paris, France
| | - Joel LeMaoult
- Commissariat ã l'Energie Atomique et aux Energies Alternatives, DRF, Francois Jacob Institute of Biology, Hemato-Immunology Research Department, Saint-Louis Hospital, Paris, France.,Université de Paris, UMR-S 976 HIPI Unit, Paris, France
| | - Nicolas O Fortunel
- Commissariat ã l'Energie Atomique et aux Energies Alternatives, DRF, Francois Jacob Institute of Biology, Laboratory of Genomics and Radiobiology of Keratinopoiesis, Institute of Cellular and Molecular Radiobiology, Evry, France.,Université Paris-Saclay, Saint-Aubin, France
| | - Michele T Martin
- Commissariat ã l'Energie Atomique et aux Energies Alternatives, DRF, Francois Jacob Institute of Biology, Laboratory of Genomics and Radiobiology of Keratinopoiesis, Institute of Cellular and Molecular Radiobiology, Evry, France.,Université Paris-Saclay, Saint-Aubin, France
| |
Collapse
|
27
|
A chalcone derivative suppresses TSLP induction in mice and human keratinocytes through binding to BET family proteins. Biochem Pharmacol 2021; 194:114819. [PMID: 34757034 DOI: 10.1016/j.bcp.2021.114819] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 10/26/2021] [Accepted: 10/27/2021] [Indexed: 11/23/2022]
Abstract
Although treatments for allergic diseases have improved, side effects and treatment resistance remain as challenges. New therapeutic drugs for allergic diseases are urgently required. Thymic stromal lymphopoietin (TSLP) is a cytokine target for prevention and treatment of allergic diseases. Since TSLP is produced from epithelial cells in allergic diseases, TSLP inhibitors may be new anti-allergic drugs. We previously identified a new inhibitor of TSLP production, named 16D10. However, its target of action remained unclarified. In this study, we found proteins binding to 16D10 from 24,000 human protein arrays by AlphaScreen-based high-throughput screening and identified bromodomain and extra-terminal (BET) family proteins as targets. We also clarified the detailed mode of interaction between 16D10 and a BET family protein using X-ray crystallography. Furthermore, we confirmed that inhibitors of BET family proteins suppressed TSLP induction and IL-33 and IL-36γ expression in both mouse and human keratinocyte cell lines. Taken together, our findings suggest that BET family proteins are involved in the suppression of TSLP production by 16D10. These proteins can contribute to the pathology of atopic dermatitis via TSLP regulation in keratinocytes and have potential as therapeutic targets in allergic diseases.
Collapse
|
28
|
Nuclear IL-33 Plays an Important Role in IL-31‒Mediated Downregulation of FLG, Keratin 1, and Keratin 10 by Regulating Signal Transducer and Activator of Transcription 3 Activation in Human Keratinocytes. J Invest Dermatol 2021; 142:136-144.e3. [PMID: 34293350 DOI: 10.1016/j.jid.2021.05.033] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 04/26/2021] [Accepted: 05/17/2021] [Indexed: 11/24/2022]
Abstract
IL-33, a chromatin-associated multifunctional cytokine, is implicated in the pathogenesis of atopic dermatitis (AD), an inflammatory skin disorder characterized by skin barrier dysfunction. IL-33 accumulates in the nuclei of epidermal keratinocytes (KCs) in AD lesions. However, it is unclear whether nuclear IL-33 directly contributes to the pathogenesis of AD. IL-31, a pruritogenic cytokine primarily produced by T helper type 2 cells, is elevated in AD lesions and promotes AD development by suppressing KC differentiation and inducing itching. In this study, we investigated the involvement of nuclear IL-33 in IL-31‒mediated suppression of KC differentiation. In monolayer cultures and living skin equivalent, IL-31 increased the expression of full-length IL-33 and the phosphorylation of signal transducer and activator of transcription 3 (STAT3) in the nuclei of human KCs, which in turn downregulated the expression of differentiation markers. We found that IL-31 and IL-4/IL-13 use very similar mechanisms to inhibit KC differentiation: nuclear IL-33 combines with phosphorylated STAT3 and functions as a STAT3 transcription cofactor, promoting phosphorylated STAT3 binding to the FLG promoter to inhibit its transcription; moreover, the nuclear IL-33/phosphorylated STAT3 complex drives the downregulation of keratin 1 and keratin 10 by reducing the availability of the transcription factor RunX1. Therefore, nuclear IL-33 plays an important role in IL-31‒mediated differentiation suppression by regulating STAT3 activation in human KCs.
Collapse
|