1
|
Maskey AR, Mo X, Li XM. Preclinical Models of Atopic Dermatitis Suitable for Mechanistic and Therapeutic Investigations. J Inflamm Res 2024; 17:6955-6970. [PMID: 39372589 PMCID: PMC11456296 DOI: 10.2147/jir.s467327] [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: 03/17/2024] [Accepted: 08/07/2024] [Indexed: 10/08/2024] Open
Abstract
Atopic dermatitis (AD) is a complex immune-mediated abnormality of the skin characterized by impaired barrier function, eczematous dermatitis, chronic pruritus and itch. The immunological response in AD is mediated by a Th2-dominated immune response in the early acute phase followed by a Th1/ Th2 mixed immune response in the chronic phase. AD is the first step of the "atopic march" that progresses into food allergy, allergic rhinitis, and asthma. Different models are indispensable for studying AD pathogenesis and for designing pre-clinical studies for therapeutic discovery. They reflect the characteristic morphological features of typical human AD with regard to epidermal thickening, hyperkeratosis, acanthosis, and spongiosis and help understand the immunopathogenesis of the disease with respect to IgE levels and cellular infiltration of eosinophils, mast cells, and lymphocytes. Although it is difficult to replicate all human AD clinical features in a model, several AD in vivo models comprising spontaneous, induced, transgenic, and humanized and in vitro models, including 2D, co-culture, and 3D, have been described previously. However, several questions remain regarding whether these models satisfactorily reflect the complexity of human AD. Therefore, this review comprehensively highlights the diversity of currently available models and provides insights into the selection of suitable models based on research questions. It also summarizes the diverse mechanisms associated with each model, which may be valuable for better study design to test new therapeutic options.
Collapse
Affiliation(s)
- Anish R Maskey
- Department of Pathology, Microbiology and Immunology, New York Medical College, Valhalla, NY, 10595, USA
| | - Xian Mo
- Department of Pathology, Microbiology and Immunology, New York Medical College, Valhalla, NY, 10595, USA
- The Department of Allergy and Clinical Immunology, Guangzhou Institute of Respiratory Health, Guangzhou, People’s Republic of China
| | - Xiu-Min Li
- Department of Pathology, Microbiology and Immunology, New York Medical College, Valhalla, NY, 10595, USA
- Department of Otolaryngology, New York Medical College, Valhalla, NY, 10595, USA
- Department of Dermatology, New York Medical College, Valhalla, NY, 10595, USA
| |
Collapse
|
2
|
Kline SN, Saito Y, Archer NK. Staphylococcus aureus Proteases: Orchestrators of Skin Inflammation. DNA Cell Biol 2024; 43:483-491. [PMID: 38957987 PMCID: PMC11535466 DOI: 10.1089/dna.2024.0134] [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: 06/07/2024] [Accepted: 06/07/2024] [Indexed: 07/04/2024] Open
Abstract
Skin homeostasis relies on a delicate balance between host proteases and protease inhibitors along with those secreted from microbial communities, as disruption to this harmony contributes to the pathogenesis of inflammatory skin disorders, including atopic dermatitis and Netherton's syndrome. In addition to being a prominent cause of skin and soft tissue infections, the gram-positive bacterium Staphylococcus aureus is a key player in inflammatory skin conditions due to its array of 10 secreted proteases. Herein we review how S. aureus proteases augment the development of inflammation in skin disorders. These mechanisms include degradation of skin barrier integrity, immune dysregulation and pruritis, and impairment of host defenses. Delineating the diverse roles of S. aureus proteases has the potential to reveal novel therapeutic strategies, such as inhibitors of proteases or their cognate target, as well as neutralizing vaccines to alleviate the burden of inflammatory skin disorders in patients.
Collapse
Affiliation(s)
- Sabrina N. Kline
- Department of Dermatology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Yoshine Saito
- School of Medicine, University of Maryland, Baltimore, Maryland, USA
| | - Nathan K. Archer
- Department of Dermatology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
3
|
Sugiura K, Fujita H, Komine M, Yamanaka K, Akiyama M. The role of interleukin-36 in health and disease states. J Eur Acad Dermatol Venereol 2024; 38:1910-1925. [PMID: 38779986 DOI: 10.1111/jdv.19935] [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: 11/03/2023] [Accepted: 01/29/2024] [Indexed: 05/25/2024]
Abstract
The interleukin (IL)-1 superfamily upregulates immune responses and maintains homeostasis between the innate and adaptive immune systems. Within the IL-1 superfamily, IL-36 plays a pivotal role in both innate and adaptive immune responses. Of the four IL-36 isoforms, three have agonist activity (IL-36α, IL-36β, IL-36γ) and the fourth has antagonist activity (IL-36 receptor antagonist [IL-36Ra]). All IL-36 isoforms bind to the IL-36 receptor (IL-36R). Binding of IL-36α/β/γ to the IL-36R recruits the IL-1 receptor accessory protein (IL-1RAcP) and activates downstream signalling pathways mediated by nuclear transcription factor kappa B and mitogen-activated protein kinase signalling pathways. Antagonist binding of IL-36Ra to IL-36R inhibits recruitment of IL-1RAcP, blocking downstream signalling pathways. Changes in the balance within the IL-36 cytokine family can lead to uncontrolled inflammatory responses throughout the body. As such, IL-36 has been implicated in numerous inflammatory diseases, notably a type of pustular psoriasis called generalized pustular psoriasis (GPP), a chronic, rare, potentially life-threatening, multisystemic skin disease characterised by recurrent fever and extensive sterile pustules. In GPP, IL-36 is central to disease pathogenesis, and the prevention of IL-36-mediated signalling can improve clinical outcomes. In this review, we summarize the literature describing the biological functions of the IL-36 pathway. We also consider the evidence for uncontrolled activation of the IL-36 pathway in a wide range of skin (e.g., plaque psoriasis, pustular psoriasis, hidradenitis suppurativa, acne, Netherton syndrome, atopic dermatitis and pyoderma gangrenosum), lung (e.g., idiopathic pulmonary fibrosis), gut (e.g., intestinal fibrosis, inflammatory bowel disease and Hirschsprung's disease), kidney (e.g., renal tubulointerstitial lesions) and infectious diseases caused by a variety of pathogens (e.g., COVID-19; Mycobacterium tuberculosis, Pseudomonas aeruginosa, Streptococcus pneumoniae infections), as well as in cancer. We also consider how targeting the IL-36 signalling pathway could be used in treating inflammatory disease states.
Collapse
Affiliation(s)
- Kazumitsu Sugiura
- Department of Dermatology, Fujita Health University School of Medicine, Toyoake, Japan
| | - Hideki Fujita
- Department of Dermatology, Nihon University School of Medicine, Tokyo, Japan
| | - Mayumi Komine
- Department of Dermatology, Faculty of Medicine, Jichi Medical University, Tochigi, Japan
| | - Keiichi Yamanaka
- Department of Dermatology, Graduate School of Medicine, Mie University, Tsu, Japan
| | - Masashi Akiyama
- Department of Dermatology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| |
Collapse
|
4
|
Keller J, O' Siorain JR, Kündig TM, Mellett M. Molecular aspects of Interleukin-36 cytokine activation and regulation. Biochem Soc Trans 2024; 52:1591-1604. [PMID: 38940747 DOI: 10.1042/bst20230548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 06/04/2024] [Accepted: 06/07/2024] [Indexed: 06/29/2024]
Abstract
Interleukin-36 (IL-36) cytokines are structurally similar to other Interleukin-1 superfamily members and are essential to convey inflammatory responses at epithelial barriers including the skin, lung, and gut. Due to their potent effects on immune cells, IL-36 cytokine activation is regulated on multiple levels, from expression and activation to receptor binding. Different IL-36 isoforms convey specific responses as a consequence of particular danger- or pathogen-associated molecular patterns. IL-36 expression and activation are regulated by exogenous pathogens, including fungi, viruses and bacteria but also by endogenous factors such as antimicrobial peptides or cytokines. Processing of IL-36 into potent bioactive forms is necessary for host protection but can elevate tissue damage. Indeed, exacerbated IL-36 signalling and hyperactivation are linked to the pathogenesis of diseases such as plaque and pustular psoriasis, emphasising the importance of understanding the molecular aspects regulating IL-36 activation. Here, we summarise facets of the electrochemical properties, regulation of extracellular cleavage by various proteases and receptor signalling of the pro-inflammatory and anti-inflammatory IL-36 family members. Additionally, this intriguing cytokine subfamily displays many characteristics that are unique from prototypical members of the IL-1 family and these key distinctions are outlined here.
Collapse
Affiliation(s)
- Jennifer Keller
- Department of Dermatology, University Hospital Zürich (USZ), University of Zürich (UZH), Raemistrasse 100, 8091 Zürich, Switzerland
- Faculty of Science, University of Zürich, 8091 Zürich, Switzerland
| | - James R O' Siorain
- Department of Dermatology, University Hospital Zürich (USZ), University of Zürich (UZH), Raemistrasse 100, 8091 Zürich, Switzerland
- Faculty of Medicine, University of Zürich, 8091 Zürich, Switzerland
| | - Thomas M Kündig
- Department of Dermatology, University Hospital Zürich (USZ), University of Zürich (UZH), Raemistrasse 100, 8091 Zürich, Switzerland
- Faculty of Medicine, University of Zürich, 8091 Zürich, Switzerland
| | - Mark Mellett
- Department of Dermatology, University Hospital Zürich (USZ), University of Zürich (UZH), Raemistrasse 100, 8091 Zürich, Switzerland
- Faculty of Medicine, University of Zürich, 8091 Zürich, Switzerland
| |
Collapse
|
5
|
Braun C, Badiou C, Guironnet-Paquet A, Iwata M, Lenief V, Mosnier A, Beauclair C, Renucci E, Bouschon P, Cuzin R, Briend Y, Patra V, Patot S, Scharschmidt TC, van Wamel W, Lemmens N, Nakajima S, Vandenesh F, Nicolas JF, Lina G, Nosbaum A, Vocanson M. Staphylococcus aureus-specific skin resident memory T cells protect against bacteria colonization but exacerbate atopic dermatitis-like flares in mice. J Allergy Clin Immunol 2024; 154:355-374. [PMID: 38734386 DOI: 10.1016/j.jaci.2024.03.032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 02/15/2024] [Accepted: 03/26/2024] [Indexed: 05/13/2024]
Abstract
BACKGROUND The contribution of Staphylococcus aureus to the exacerbation of atopic dermatitis (AD) is widely documented, but its role as a primary trigger of AD skin symptoms remains poorly explored. OBJECTIVES This study sought to reappraise the main bacterial factors and underlying immune mechanisms by which S aureus triggers AD-like inflammation. METHODS This study capitalized on a preclinical model, in which different clinical isolates were applied in the absence of any prior experimental skin injury. RESULTS The development of S aureus-induced dermatitis depended on the nature of the S aureus strain, its viability, the concentration of the applied bacterial suspension, the production of secreted and nonsecreted factors, as well as the activation of accessory gene regulatory quorum sensing system. In addition, the rising dermatitis, which exhibited the well-documented AD cytokine signature, was significantly inhibited in inflammasome adaptor apoptosis-associated speck-like protein containing a CARD domain- and monocyte/macrophage-deficient animals, but not in T- and B-cell-deficient mice, suggesting a major role for the innate response in the induction of skin inflammation. However, bacterial exposure generated a robust adaptive immune response against S aureus, and an accumulation of S aureus-specific γδ and CD4+ tissue resident memory T cells at the site of previous dermatitis. The latter both contributed to worsen the flares of AD-like dermatitis on new bacteria exposures, but also, protected the mice from persistent bacterial colonization. CONCLUSIONS These data highlight the induction of unique AD-like inflammation, with the generation of proinflammatory but protective tissue resident memory T cells in a context of natural exposure to pathogenic S aureus strains.
Collapse
Affiliation(s)
- Camille Braun
- Centre International de Recherche en Infectiologie (CIRI), INSERM U1111, Unité Mixte de Recherche 5308, Centre national de la recherche scientifique, Ecole Normale Supérieure de Lyon, Lyon, France; Service de Pédiatrie, Pneumologie, Allergologie, Mucoviscidose, Hôpital Femme Mère Enfant, Hospices Civils de Lyon, Bron, France
| | - Cédric Badiou
- Centre International de Recherche en Infectiologie (CIRI), INSERM U1111, Unité Mixte de Recherche 5308, Centre national de la recherche scientifique, Ecole Normale Supérieure de Lyon, Lyon, France
| | - Aurélie Guironnet-Paquet
- Centre International de Recherche en Infectiologie (CIRI), INSERM U1111, Unité Mixte de Recherche 5308, Centre national de la recherche scientifique, Ecole Normale Supérieure de Lyon, Lyon, France; Etablissement Français du Sang Auvergne Rhône-Alpes, Apheresis Unit, Hôpital Lyon Sud, Pierre-Bénite, France
| | - Masashi Iwata
- Department of Dermatology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Vanina Lenief
- Centre International de Recherche en Infectiologie (CIRI), INSERM U1111, Unité Mixte de Recherche 5308, Centre national de la recherche scientifique, Ecole Normale Supérieure de Lyon, Lyon, France
| | - Amandine Mosnier
- Centre International de Recherche en Infectiologie (CIRI), INSERM U1111, Unité Mixte de Recherche 5308, Centre national de la recherche scientifique, Ecole Normale Supérieure de Lyon, Lyon, France
| | - Charlotte Beauclair
- Centre International de Recherche en Infectiologie (CIRI), INSERM U1111, Unité Mixte de Recherche 5308, Centre national de la recherche scientifique, Ecole Normale Supérieure de Lyon, Lyon, France
| | - Emilie Renucci
- Centre International de Recherche en Infectiologie (CIRI), INSERM U1111, Unité Mixte de Recherche 5308, Centre national de la recherche scientifique, Ecole Normale Supérieure de Lyon, Lyon, France
| | - Pauline Bouschon
- Centre International de Recherche en Infectiologie (CIRI), INSERM U1111, Unité Mixte de Recherche 5308, Centre national de la recherche scientifique, Ecole Normale Supérieure de Lyon, Lyon, France
| | - Roxane Cuzin
- Centre International de Recherche en Infectiologie (CIRI), INSERM U1111, Unité Mixte de Recherche 5308, Centre national de la recherche scientifique, Ecole Normale Supérieure de Lyon, Lyon, France
| | - Yoann Briend
- Centre International de Recherche en Infectiologie (CIRI), INSERM U1111, Unité Mixte de Recherche 5308, Centre national de la recherche scientifique, Ecole Normale Supérieure de Lyon, Lyon, France
| | - Vijaykumar Patra
- Centre International de Recherche en Infectiologie (CIRI), INSERM U1111, Unité Mixte de Recherche 5308, Centre national de la recherche scientifique, Ecole Normale Supérieure de Lyon, Lyon, France
| | - Sabine Patot
- Centre International de Recherche en Infectiologie (CIRI), INSERM U1111, Unité Mixte de Recherche 5308, Centre national de la recherche scientifique, Ecole Normale Supérieure de Lyon, Lyon, France
| | | | - Willem van Wamel
- Department of Medical Microbiology and Infectious Diseases, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Nicole Lemmens
- Department of Medical Microbiology and Infectious Diseases, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Saeko Nakajima
- Department of Dermatology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - François Vandenesh
- Centre International de Recherche en Infectiologie (CIRI), INSERM U1111, Unité Mixte de Recherche 5308, Centre national de la recherche scientifique, Ecole Normale Supérieure de Lyon, Lyon, France; Service de Microbiologie Clinique, Groupement Hospitalier Nord, Hospices Civils de Lyon, Bron, France; Centre National de Référence des Staphylocoques, Hospices Civils de Lyon, Lyon, France
| | - Jean-François Nicolas
- Centre International de Recherche en Infectiologie (CIRI), INSERM U1111, Unité Mixte de Recherche 5308, Centre national de la recherche scientifique, Ecole Normale Supérieure de Lyon, Lyon, France; Service d'Allergologie et Immunologie Clinique, Centre Hospitalier Lyon Sud, Hospices Civils de Lyon, Pierre-Bénite, France
| | - Gérard Lina
- Centre International de Recherche en Infectiologie (CIRI), INSERM U1111, Unité Mixte de Recherche 5308, Centre national de la recherche scientifique, Ecole Normale Supérieure de Lyon, Lyon, France; Service de Microbiologie Clinique, Groupement Hospitalier Nord, Hospices Civils de Lyon, Bron, France; Centre National de Référence des Staphylocoques, Hospices Civils de Lyon, Lyon, France
| | - Audrey Nosbaum
- Centre International de Recherche en Infectiologie (CIRI), INSERM U1111, Unité Mixte de Recherche 5308, Centre national de la recherche scientifique, Ecole Normale Supérieure de Lyon, Lyon, France; Service d'Allergologie et Immunologie Clinique, Centre Hospitalier Lyon Sud, Hospices Civils de Lyon, Pierre-Bénite, France
| | - Marc Vocanson
- Centre International de Recherche en Infectiologie (CIRI), INSERM U1111, Unité Mixte de Recherche 5308, Centre national de la recherche scientifique, Ecole Normale Supérieure de Lyon, Lyon, France.
| |
Collapse
|
6
|
Yousef A. Alnakeeb E, Abdull Razis AF, Wei Chan K, Ling Tham C, Han Chan Y, Kalifa Kafo AS, Jambari NN, Rollin P, Djedaini-Pilard F. Antiallergic Effect of the Alpha-Cyclodextrin Moringin Complex in Rat Basophilic Leukaemia (RBL-2H3) Cell Line. Adv Pharmacol Pharm Sci 2024; 2024:8885068. [PMID: 39104427 PMCID: PMC11300081 DOI: 10.1155/2024/8885068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 03/19/2024] [Accepted: 06/25/2024] [Indexed: 08/07/2024] Open
Abstract
Allergic diseases (ADs) are a major concern when it comes to public well-being. Moringa oleifera Lam is a tropical plant that is used in traditional medicine due to the presence of isothiocyanate. The present study investigated the antiallergic properties of 4-(α-L-rhamnopyranosyloxy)-benzyl isothiocyanate or moringin isolated from Moringa oleifera seeds in the form of alpha-cyclodextrin-moringin (α-CD/MG) complex on rat basophilic leukaemia (RBL-2H3) cell line at both the early and late stages of an allergic reaction. The α-CD/MG complex was initially elucidated using nuclear magnetic resonance (NMR) followed by the 3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium inner salt proliferation assay to evaluate the cytotoxicity and cell viability with respect to ketotifen fumarate (KF) and α-CD/MG. The release of beta-hexosaminidase (β-hexosaminidase) and histamine was used to determine the level of inhibition in the early stage while the suppression of the release of prostaglandin (PGD2), tumour necrosis factor-alpha (TNF-α), and interleukin (IL-4) was considered in the late stage. Higher concentrations of α-CD/MG (5 μM, p < 0.001) in mast cell degranulation significantly inhibited the expression of β-hexosaminidase, histamine, TNF-α, PGD2, and IL-4 in both the early and late stages. Thus, α-CD/MG can potentially be developed as an antiallergic drug as it has the ability to inhibit allergic responses in the late and early stages.
Collapse
Affiliation(s)
- Ebtisam Yousef A. Alnakeeb
- Natural Medicines and Products Research LaboratoryInstitute of BioscienceUniversiti Putra Malaysia, UPM Serdang 43400, Selangor, Malaysia
| | - Ahmad Faizal Abdull Razis
- Natural Medicines and Products Research LaboratoryInstitute of BioscienceUniversiti Putra Malaysia, UPM Serdang 43400, Selangor, Malaysia
- Laboratory of Food Safety and Food IntegrityInstitute of Tropical Agriculture and Food SecurityUniversiti Putra Malaysia, UPM Serdang 43400, Selangor, Malaysia
- Department of Food ScienceFaculty of Food Science and TechnologyUniversiti Putra Malaysia, UPM Serdang 43400, Selangor, Malaysia
| | - Kim Wei Chan
- Natural Medicines and Products Research LaboratoryInstitute of BioscienceUniversiti Putra Malaysia, UPM Serdang 43400, Selangor, Malaysia
| | - Chau Ling Tham
- Department of Biomedical SciencesFaculty of Medicine and Health SciencesUniversiti Putra Malaysia, UPM Serdang 43400, Selangor, Malaysia
| | - Yee Han Chan
- Department of Biomedical SciencesFaculty of Medicine and Health SciencesUniversiti Putra Malaysia, UPM Serdang 43400, Selangor, Malaysia
| | - Anwar Salm Kalifa Kafo
- Department of PathologyFaculty of Medicine and Health SciencesUniversiti Putra Malaysia, UPM Serdang 43400, Selangor, Malaysia
| | - Nuzul Noorahya Jambari
- Laboratory of Food Safety and Food IntegrityInstitute of Tropical Agriculture and Food SecurityUniversiti Putra Malaysia, UPM Serdang 43400, Selangor, Malaysia
- Department of Food ScienceFaculty of Food Science and TechnologyUniversiti Putra Malaysia, UPM Serdang 43400, Selangor, Malaysia
| | - Patrick Rollin
- Université d'Orléans et CNRSICOA, UMR 7311, BP 6759, CEDEX 02, Orléans F-45067, France
| | - Florence Djedaini-Pilard
- LG2A UR 7378Université de Picardie Jules Verne33 rue Saint Leu—UFR des Sciences, Amiens F-80000, France
| |
Collapse
|
7
|
Yang XP, Liu YY, Zhang CY, Huang KK, Han SS, Liang BY, Lin Y. An Observational Study: Association Between Atopic Dermatitis and Bacterial Colony of the Skin Based on 16S rRNA Gene Sequencing. Clin Cosmet Investig Dermatol 2024; 17:1649-1659. [PMID: 39050561 PMCID: PMC11268436 DOI: 10.2147/ccid.s464431] [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: 04/09/2024] [Accepted: 06/23/2024] [Indexed: 07/27/2024]
Abstract
Aim Atopic dermatitis (AD) often accompanies skin infections, and bacterial skin infections often cause persistent and worsening symptoms. In this study, we explored the key changes in the microbiota of AD patients, as well as the effects of different ages and the severity of rash on changes in the microbiota. Patients and Methods A total of 95 AD patients and 77 healthy volunteers were recruited. The AD patients were divided into three groups based age and three groups according to the EASI score. Microorganisms collected from the skin were analyzed through 16S rRNA gene sequencing, revealing species diversity via α and β diversity analyses. Species compositions were compared at the phylum and genus levels. The significance of skin microbiota at the genus level was assessed using the random forest algorithm. Finally, the impact of relationships between different microbial communities on the microbial community composition and the pathogenesis of AD was explored using Pearson correlation coefficients. Results The species diversity of the skin microbiota in the AD group significantly decreased. Compared with that in the healthy volunteers (HV) group, the bacterial diversity in the two groups of samples significantly differed. Staphylococcus dominated the bacterial communities, and as AD symptoms gradually worsened, the abundance of Staphylococcus gradually increased. Among all bacterial genera with a relative abundance greater than 1%, Staphylococcus showed a negative correlation with other genera, and showed significant consistency in specimens from different age groups. Conclusion Changes in the abundance of Staphylococcus in the skin bacterial colonies are the main cause of AD. Brevundimonas, Paracoccus, Corynebacterium, and Veillonella may serve as characteristic biomarkers for AD. These results indicate that altering the microbiota composition of the skin may aid in the treatment of AD.
Collapse
Affiliation(s)
- Xian-Ping Yang
- Department of Dermatology, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou, Guangdong, People’s Republic of China
| | - Ying-Yao Liu
- The Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, People’s Republic of China
| | - Cai-Yun Zhang
- Department of Dermatology, Dongguan Traditional Chinese Medicine Hospital, Dongguan, Guangdong, People’s Republic of China
| | - Kai-Kai Huang
- Department of Dermatology, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou, Guangdong, People’s Republic of China
| | - Shan-Shan Han
- Department of Dermatology, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou, Guangdong, People’s Republic of China
| | - Bao-Ying Liang
- Department of Dermatology, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou, Guangdong, People’s Republic of China
| | - Ying Lin
- Department of Dermatology, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou, Guangdong, People’s Republic of China
| |
Collapse
|
8
|
Zhang S, Fang X, Xu B, Zhou Y, Li F, Gao Y, Luo Y, Yao X, Liu X. Comprehensive analysis of phenotypes and transcriptome characteristics reveal the best atopic dermatitis mouse model induced by MC903. J Dermatol Sci 2024; 114:104-114. [PMID: 38806322 DOI: 10.1016/j.jdermsci.2024.05.003] [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: 01/05/2024] [Revised: 04/01/2024] [Accepted: 05/13/2024] [Indexed: 05/30/2024]
Abstract
BACKGROUND Although several mouse models of exogenous-agent-induced atopic dermatitis (AD) are currently available, the lack of certainty regarding their similarity with human AD has limited their scientific value. Thus, comprehensive evaluation of the characteristics of mouse models and their similarity with human AD is essential. OBJECTIVE To compare six different exogenous-agent-induced AD mouse models and find out the optimum models for study. METHODS Female BALB/c mice underwent induction of AD-like dermatitis by MC903 alone or in combination with ovalbumin (OVA), dinitrofluorobenzene (DNFB) alone or in combination with OVA, OVA alone, or Staphylococcus aureus. Gross phenotype, total immunoglobulin E (IgE) level, histopathological manifestations, and skin lesion transcriptome were analyzed, and metagenomic sequencing of the gut microbiome was performed. RESULTS The DNFB plus OVA model showed the highest disease severity, while the OVA model showed the lowest severity. The MC903 and MC903 plus OVA models showed high expression of T-helper (Th)2- and Th17-related genes; the DNFB and DNFB plus OVA models showed upregulation of Th1-, Th2-, and Th17-related genes; while the S. aureus inoculation model showed more enhanced Th1 and Th17 immune responses. In contrast to the other models, the OVA-induced model showed the lowest expression levels of inflammation-related genes, while the MC903 model shared the largest overlap with human AD profiles. The intestinal microbiota of all groups showed significant differences after modeling. CONCLUSION Each AD mouse model exhibited different characteristics. The MC903 model was the best to recapitulate most features of human AD among these exogenous-agent-induced AD models.
Collapse
Affiliation(s)
- Shan Zhang
- Department of Allergy and Rheumatology, Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences & Peking Union Medical College, Nanjing, China
| | - Xiaokai Fang
- Department of Allergy and Rheumatology, Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences & Peking Union Medical College, Nanjing, China
| | - Beilei Xu
- Department of Allergy and Rheumatology, Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences & Peking Union Medical College, Nanjing, China
| | - Yuan Zhou
- Department of Allergy and Rheumatology, Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences & Peking Union Medical College, Nanjing, China
| | - Fang Li
- Department of Allergy and Rheumatology, Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences & Peking Union Medical College, Nanjing, China
| | - Yuwen Gao
- Department of Allergy and Rheumatology, Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences & Peking Union Medical College, Nanjing, China
| | - Yang Luo
- Department of Allergy and Rheumatology, Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences & Peking Union Medical College, Nanjing, China
| | - Xu Yao
- Department of Allergy and Rheumatology, Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences & Peking Union Medical College, Nanjing, China.
| | - Xiaochun Liu
- Department of Allergy and Rheumatology, Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences & Peking Union Medical College, Nanjing, China.
| |
Collapse
|
9
|
Tongmuang N, Cai KQ, An J, Novy M, Jensen LE. Floxed Il1rl2 Locus with mCherry Reporter Element Reveals Distinct Expression Patterns of the IL-36 Receptor in Barrier Tissues. Cells 2024; 13:787. [PMID: 38727323 PMCID: PMC11083296 DOI: 10.3390/cells13090787] [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: 04/09/2024] [Revised: 04/30/2024] [Accepted: 05/02/2024] [Indexed: 05/13/2024] Open
Abstract
IL-36 cytokines are emerging as beneficial in immunity against pathogens and cancers but can also be detrimental when dysregulated in autoimmune and autoinflammatory conditions. Interest in targeting IL-36 activity for therapeutic purposes is rapidly growing, yet many unknowns about the functions of these cytokines remain. Thus, the availability of robust research tools is essential for both fundamental basic science and pre-clinical studies to fully access outcomes of any manipulation of the system. For this purpose, a floxed Il1rl2, the gene encoding the IL-36 receptor, mouse strain was developed to facilitate the generation of conditional knockout mice. The targeted locus was engineered to contain an inverted mCherry reporter sequence that upon Cre-mediated recombination will be flipped and expressed under the control of the endogenous Il1rl2 promoter. This feature can be used to confirm knockout in individual cells but also as a reporter to determine which cells express the IL-36 receptor IL-1RL2. The locus was confirmed to function as intended and further used to demonstrate the expression of IL-1RL2 in barrier tissues. Il1rl2 expression was detected in leukocytes in all barrier tissues. Interestingly, strong expression was observed in epithelial cells at locations in direct contact with the environment such as the skin, oral mucosa, the esophagus, and the upper airways, but almost absent from epithelial cells at more inward facing sites, including lung alveoli, the small intestine, and the colon. These findings suggest specialized functions of IL-1RL2 in outward facing epithelial tissues and cells. The generated mouse model should prove valuable in defining such functions and may also facilitate basic and translational research.
Collapse
Affiliation(s)
- Nopprarat Tongmuang
- Department of Microbiology, Immunology and Inflammation, Temple University Lewis Katz School of Medicine, Philadelphia, PA 19140, USA
- Center for Inflammation and Lung Research, Temple University Lewis Katz School of Medicine, Philadelphia, PA 19140, USA
| | - Kathy Q. Cai
- Histopathology Facility, Fox Chase Cancer Center, Temple Health, Philadelphia, PA 19111, USA
| | - Jiahui An
- Department of Microbiology, Immunology and Inflammation, Temple University Lewis Katz School of Medicine, Philadelphia, PA 19140, USA
- Center for Inflammation and Lung Research, Temple University Lewis Katz School of Medicine, Philadelphia, PA 19140, USA
| | - Mariah Novy
- Department of Microbiology, Immunology and Inflammation, Temple University Lewis Katz School of Medicine, Philadelphia, PA 19140, USA
- Center for Inflammation and Lung Research, Temple University Lewis Katz School of Medicine, Philadelphia, PA 19140, USA
| | - Liselotte E. Jensen
- Department of Microbiology, Immunology and Inflammation, Temple University Lewis Katz School of Medicine, Philadelphia, PA 19140, USA
- Center for Inflammation and Lung Research, Temple University Lewis Katz School of Medicine, Philadelphia, PA 19140, USA
- Cancer Signaling and Microenvironment, Fox Chase Cancer Center, Temple Health, Philadelphia, PA 19111, USA
| |
Collapse
|
10
|
Schuler CF, Tsoi LC, Billi AC, Harms PW, Weidinger S, Gudjonsson JE. Genetic and Immunological Pathogenesis of Atopic Dermatitis. J Invest Dermatol 2024; 144:954-968. [PMID: 38085213 PMCID: PMC11040454 DOI: 10.1016/j.jid.2023.10.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 10/05/2023] [Accepted: 10/25/2023] [Indexed: 02/03/2024]
Abstract
Type 2 immune-mediated diseases give a clear answer to the issue of nature (genetics) versus nurture (environment). Both genetics and environment play vital complementary roles in the development of atopic dermatitis (AD). As a key component of the atopic march, AD demonstrates the interactive nature of genetic and environmental contributions to atopy. From sequence variants in the epithelial barrier gene encoding FLG to the hygiene hypothesis, AD combines a broad array of contributions into a single syndrome. This review will focus on the genetic contribution to AD and where genetics facilitates the elicitation or enhancement of AD pathogenesis.
Collapse
Affiliation(s)
- Charles F Schuler
- Division of Allergy and Clinical Immunology, Department of Internal Medicine, Michigan Medicine, University of Michigan, Ann Arbor, Michigan, USA; Mary H. Weiser Food Allergy Center, Michigan Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Lam C Tsoi
- Mary H. Weiser Food Allergy Center, Michigan Medicine, University of Michigan, Ann Arbor, Michigan, USA; Department of Dermatology, Michigan Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Allison C Billi
- Department of Dermatology, Michigan Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Paul W Harms
- Department of Dermatology, Michigan Medicine, University of Michigan, Ann Arbor, Michigan, USA; Department of Pathology, Michigan Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Stephan Weidinger
- Department of Dermatology, Venereology, and Allergology, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Johann E Gudjonsson
- Mary H. Weiser Food Allergy Center, Michigan Medicine, University of Michigan, Ann Arbor, Michigan, USA; Department of Dermatology, Michigan Medicine, University of Michigan, Ann Arbor, Michigan, USA.
| |
Collapse
|
11
|
Surendar J, Hackenberg RK, Schmitt-Sánchez F, Ossendorff R, Welle K, Stoffel-Wagner B, Sage PT, Burger C, Wirtz DC, Strauss AC, Schildberg FA. Osteomyelitis is associated with increased anti-inflammatory response and immune exhaustion. Front Immunol 2024; 15:1396592. [PMID: 38736874 PMCID: PMC11082283 DOI: 10.3389/fimmu.2024.1396592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Accepted: 04/12/2024] [Indexed: 05/14/2024] Open
Abstract
Introduction Osteomyelitis (OMS) is a bone infection causing bone pain and severe complications. A balanced immune response is critical to eradicate infection without harming the host, yet pathogens manipulate immunity to establish a chronic infection. Understanding OMS-driven inflammation is essential for disease management, but comprehensive data on immune profiles and immune cell activation during OMS are lacking. Methods Using high-dimensional flow cytometry, we investigated the detailed innate and adaptive systemic immune cell populations in OMS and age- and sex-matched controls. Results Our study revealed that OMS is associated with increased levels of immune regulatory cells, namely T regulatory cells, B regulatory cells, and T follicular regulatory cells. In addition, the expression of immune activation markers HLA-DR and CD86 was decreased in OMS, while the expression of immune exhaustion markers TIM-3, PD-1, PD-L1, and VISTA was increased. Members of the T follicular helper (Tfh) cell family as well as classical and typical memory B cells were significantly increased in OMS individuals. We also found a strong correlation between memory B cells and Tfh cells. Discussion We conclude that OMS skews the host immune system towards the immunomodulatory arm and that the Tfh memory B cell axis is evident in OMS. Therefore, immune-directed therapies may be a promising alternative for eradication and recurrence of infection in OMS, particularly in individuals and areas where antibiotic resistance is a major concern.
Collapse
Affiliation(s)
- Jayagopi Surendar
- Department of Orthopedics and Trauma Surgery, University Hospital Bonn, Bonn, Germany
| | - Roslind K. Hackenberg
- Department of Orthopedics and Trauma Surgery, University Hospital Bonn, Bonn, Germany
- Department of Hand, Plastic and Reconstructive Surgery, Burn Center, BG Trauma Center Ludwigshafen, University of Heidelberg, Ludwigshafen, Germany
| | - Fabio Schmitt-Sánchez
- Department of Orthopedics and Trauma Surgery, University Hospital Bonn, Bonn, Germany
| | - Robert Ossendorff
- Department of Orthopedics and Trauma Surgery, University Hospital Bonn, Bonn, Germany
| | - Kristian Welle
- Department of Orthopedics and Trauma Surgery, University Hospital Bonn, Bonn, Germany
| | - Birgit Stoffel-Wagner
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, Bonn, Germany
| | - Peter T. Sage
- Transplantation Research Center, Renal Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Christof Burger
- Department of Orthopedics and Trauma Surgery, University Hospital Bonn, Bonn, Germany
| | - Dieter C. Wirtz
- Department of Orthopedics and Trauma Surgery, University Hospital Bonn, Bonn, Germany
| | - Andreas C. Strauss
- Department of Orthopedics and Trauma Surgery, University Hospital Bonn, Bonn, Germany
| | - Frank A. Schildberg
- Department of Orthopedics and Trauma Surgery, University Hospital Bonn, Bonn, Germany
| |
Collapse
|
12
|
Clowry J, Dempsey DJ, Claxton TJ, Towell AM, Turley MB, Sutton M, Geoghegan JA, Kezic S, Jakasa I, White A, Irvine AD, McLoughlin RM. Distinct T cell signatures are associated with Staphylococcus aureus skin infection in pediatric atopic dermatitis. JCI Insight 2024; 9:e178789. [PMID: 38716729 PMCID: PMC11141913 DOI: 10.1172/jci.insight.178789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Accepted: 04/03/2024] [Indexed: 06/02/2024] Open
Abstract
Atopic dermatitis (AD) is an inflammatory skin condition with a childhood prevalence of up to 25%. Microbial dysbiosis is characteristic of AD, with Staphylococcus aureus the most frequent pathogen associated with disease flares and increasingly implicated in disease pathogenesis. Therapeutics to mitigate the effects of S. aureus have had limited efficacy and S. aureus-associated temporal disease flares are synonymous with AD. An alternative approach is an anti-S. aureus vaccine, tailored to AD. Experimental vaccines have highlighted the importance of T cells in conferring protective anti-S. aureus responses; however, correlates of T cell immunity against S. aureus in AD have not been identified. We identify a systemic and cutaneous immunological signature associated with S. aureus skin infection (ADS.aureus) in a pediatric AD cohort, using a combined Bayesian multinomial analysis. ADS.aureus was most highly associated with elevated cutaneous chemokines IP10 and TARC, which preferentially direct Th1 and Th2 cells to skin. Systemic CD4+ and CD8+ T cells, except for Th2 cells, were suppressed in ADS.aureus, particularly circulating Th1, memory IL-10+ T cells, and skin-homing memory Th17 cells. Systemic γδ T cell expansion in ADS.aureus was also observed. This study suggests that augmentation of protective T cell subsets is a potential therapeutic strategy in the management of S. aureus in AD.
Collapse
Affiliation(s)
- Julianne Clowry
- Department of Dermatology, National Children’s Research Centre, Children’s Health Ireland at Crumlin, Dublin, Ireland
- Clinical Medicine, Trinity College Dublin, Dublin, Ireland
- Host-Pathogen Interactions Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Daniel J. Dempsey
- Host-Pathogen Interactions Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Tracey J. Claxton
- Host-Pathogen Interactions Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Aisling M. Towell
- Department of Microbiology, Moyne Institute of Preventive Medicine, School of Genetics and Microbiology, Trinity College Dublin, Dublin, Ireland
| | - Mary B. Turley
- Department of Microbiology, Moyne Institute of Preventive Medicine, School of Genetics and Microbiology, Trinity College Dublin, Dublin, Ireland
- Institute of Microbiology and Infection, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Martin Sutton
- Department of Microbiology, Moyne Institute of Preventive Medicine, School of Genetics and Microbiology, Trinity College Dublin, Dublin, Ireland
| | - Joan A. Geoghegan
- Department of Microbiology, Moyne Institute of Preventive Medicine, School of Genetics and Microbiology, Trinity College Dublin, Dublin, Ireland
- Institute of Microbiology and Infection, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Sanja Kezic
- Amsterdam UMC, University of Amsterdam, Department of Public and Occupational Health, Amsterdam Public Health Research Institute, Amsterdam, Netherlands
| | - Ivone Jakasa
- Laboratory for Analytical Chemistry, Department of Chemistry and Biochemistry, Faculty of Food Technology and Biotechnology, University of Zagreb, Zagreb, Croatia
| | - Arthur White
- School of Computer Science and Statistics, Trinity College Dublin, Dublin, Ireland
| | - Alan D. Irvine
- Department of Dermatology, National Children’s Research Centre, Children’s Health Ireland at Crumlin, Dublin, Ireland
- Clinical Medicine, Trinity College Dublin, Dublin, Ireland
| | - Rachel M. McLoughlin
- Host-Pathogen Interactions Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
13
|
Yang Y, Huang J, Zeng A, Long X, Yu N, Wang X. The role of the skin microbiome in wound healing. BURNS & TRAUMA 2024; 12:tkad059. [PMID: 38444635 PMCID: PMC10914219 DOI: 10.1093/burnst/tkad059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/11/2023] [Revised: 08/05/2023] [Accepted: 11/21/2023] [Indexed: 03/07/2024]
Abstract
The efficient management of skin wounds for rapid and scarless healing represents a major clinical unmet need. Nonhealing skin wounds and undesired scar formation impair quality of life and result in high healthcare expenditure worldwide. The skin-colonizing microbiota contributes to maintaining an intact skin barrier in homeostasis, but it also participates in the pathogenesis of many skin disorders, including aberrant wound healing, in many respects. This review focuses on the composition of the skin microbiome in cutaneous wounds of different types (i.e. acute and chronic) and with different outcomes (i.e. nonhealing and hypertrophic scarring), mainly based on next-generation sequencing analyses; furthermore, we discuss the mechanistic insights into host-microbe and microbe-microbe interactions during wound healing. Finally, we highlight potential therapeutic strategies that target the skin microbiome to improve healing outcomes.
Collapse
Affiliation(s)
- Yuyan Yang
- Department of Plastic and Reconstructive Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 1, Shuaifuyuan, Dongcheng District, Beijing, 100005, China
| | - Jiuzuo Huang
- Department of Plastic and Reconstructive Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 1, Shuaifuyuan, Dongcheng District, Beijing, 100005, China
| | - Ang Zeng
- Department of Plastic and Reconstructive Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 1, Shuaifuyuan, Dongcheng District, Beijing, 100005, China
| | - Xiao Long
- Department of Plastic and Reconstructive Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 1, Shuaifuyuan, Dongcheng District, Beijing, 100005, China
| | - Nanze Yu
- Department of Plastic and Reconstructive Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 1, Shuaifuyuan, Dongcheng District, Beijing, 100005, China
| | - Xiaojun Wang
- Department of Plastic and Reconstructive Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 1, Shuaifuyuan, Dongcheng District, Beijing, 100005, China
| |
Collapse
|
14
|
Kline SN, Orlando NA, Lee AJ, Wu MJ, Zhang J, Youn C, Feller LE, Pontaza C, Dikeman D, Limjunyawong N, Williams KL, Wang Y, Cihakova D, Jacobsen EA, Durum SK, Garza LA, Dong X, Archer NK. Staphylococcus aureus proteases trigger eosinophil-mediated skin inflammation. Proc Natl Acad Sci U S A 2024; 121:e2309243121. [PMID: 38289950 PMCID: PMC10861893 DOI: 10.1073/pnas.2309243121] [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: 06/06/2023] [Accepted: 12/22/2023] [Indexed: 02/01/2024] Open
Abstract
Staphylococcus aureus skin colonization and eosinophil infiltration are associated with many inflammatory skin disorders, including atopic dermatitis, bullous pemphigoid, Netherton's syndrome, and prurigo nodularis. However, whether there is a relationship between S. aureus and eosinophils and how this interaction influences skin inflammation is largely undefined. We show in a preclinical mouse model that S. aureus epicutaneous exposure induced eosinophil-recruiting chemokines and eosinophil infiltration into the skin. Remarkably, we found that eosinophils had a comparable contribution to the skin inflammation as T cells, in a manner dependent on eosinophil-derived IL-17A and IL-17F production. Importantly, IL-36R signaling induced CCL7-mediated eosinophil recruitment to the inflamed skin. Last, S. aureus proteases induced IL-36α expression in keratinocytes, which promoted infiltration of IL-17-producing eosinophils. Collectively, we uncovered a mechanism for S. aureus proteases to trigger eosinophil-mediated skin inflammation, which has implications in the pathogenesis of inflammatory skin diseases.
Collapse
Affiliation(s)
- Sabrina N. Kline
- Department of Dermatology, Johns Hopkins School of Medicine, Baltimore, MD21287
| | - Nicholas A. Orlando
- Department of Dermatology, Johns Hopkins School of Medicine, Baltimore, MD21287
| | - Alex J. Lee
- Department of Oncology, Bloomberg Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD21205
| | - Meng-Jen Wu
- Department of Dermatology, Johns Hopkins School of Medicine, Baltimore, MD21287
| | - Jing Zhang
- Department of Dermatology, Johns Hopkins School of Medicine, Baltimore, MD21287
| | - Christine Youn
- Department of Dermatology, Johns Hopkins School of Medicine, Baltimore, MD21287
| | - Laine E. Feller
- Department of Dermatology, Johns Hopkins School of Medicine, Baltimore, MD21287
| | - Cristina Pontaza
- Department of Dermatology, Johns Hopkins School of Medicine, Baltimore, MD21287
| | - Dustin Dikeman
- Department of Dermatology, Johns Hopkins School of Medicine, Baltimore, MD21287
| | - Nathachit Limjunyawong
- Center of Research Excellence in Allergy and Immunology, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok10700, Thailand
| | - Kaitlin L. Williams
- Department of Dermatology, Johns Hopkins School of Medicine, Baltimore, MD21287
| | - Yu Wang
- Department of Dermatology, Johns Hopkins School of Medicine, Baltimore, MD21287
| | - Daniela Cihakova
- Department of Pathology, Johns Hopkins School of Medicine, Baltimore, MD21287
| | - Elizabeth A. Jacobsen
- Division of Allergy, Asthma and Clinical Immunology, Mayo Clinic Arizona, Scottsdale, AZ85259
| | - Scott K. Durum
- Cancer Innovation Laboratory, Center for Cancer Research, National Cancer Institute, NIH, Frederick, MD21702
| | - Luis A. Garza
- Department of Dermatology, Johns Hopkins School of Medicine, Baltimore, MD21287
| | - Xinzhong Dong
- HHMI, Johns Hopkins University School of Medicine, Baltimore, MD21205
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD21205
| | - Nathan K. Archer
- Department of Dermatology, Johns Hopkins School of Medicine, Baltimore, MD21287
| |
Collapse
|
15
|
Zhong Y, Huang T, Li X, Luo P, Zhang B. GSDMD suppresses keratinocyte differentiation by inhibiting FLG expression and attenuating KCTD6-mediated HDAC1 degradation in atopic dermatitis. PeerJ 2024; 12:e16768. [PMID: 38250727 PMCID: PMC10798152 DOI: 10.7717/peerj.16768] [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: 07/14/2023] [Accepted: 12/15/2023] [Indexed: 01/23/2024] Open
Abstract
Background Recent studies have shown that activated pyroptosis in atopic dermatitis (AD) switches inflammatory processes and causes abnormal cornification and epidermal barrier dysfunction. Little research has focused on the interaction mechanism between pyroptosis-related genes and human keratinocyte differentiation. Methods The AD dataset from the Gene Expression Omnibus (GEO) was used to identify differently expressed pyroptosis-related genes (DEPRGs). Hub genes were identified and an enrichment analysis was performed to select epithelial development-related genes. Lesions of AD patients were detected via immunohistochemistry (IHC) to verify the hub gene. Human keratinocytes cell lines, gasdermin D (GSDMD) overexpression, Caspase1 siRNA, Histone Deacetylase1 (HDAC1) siRNA, and HDAC1 overexpression vectors were used for gain-and-loss-of-function experiments. Regulation of cornification protein was determined by qPCR, western blot (WB), immunofluorescence (IF), dual-luciferase reporter assay, co-immunoprecipitation (Co-IP), and chromatin immunoprecipitation (ChIP). Results A total of 27 DEPRGs were identified between either atopic dermatitis non-lesional skin (ANL) and healthy control (HC) or atopic dermatitis lesional skin (AL) and HC. The enrichment analysis showed that these DEPRGs were primarily enriched in the inflammatory response and keratinocytes differentiation. Of the 10 hub genes identified via the protein-protein interaction network, only GSDMD was statistically and negatively associated with the expression of epithelial tight junction core genes. Furthermore, GSDMD was upregulated in AD lesions and inhibited human keratinocyte differentiation by reducing filaggrin (FLG) expression. Mechanistically, GSDMD activated by Caspase1 reduced FLG expression via HDAC1. HDAC1 decreased FLG expression by reducing histone acetylation at the FLG promoter. In addition, GSDMD blocked the interaction of Potassium Channel Tetramerization Domain Containing 6 (KCTD6) and HDAC1 to prohibit HDAC1 degradation. Conclusion This study revealed that GSDMD was upregulated in AD lesions and that GSDMD regulated keratinocytes via epigenetic modification, which might provide potential therapeutic targets for AD.
Collapse
Affiliation(s)
- Yi Zhong
- Department of Dermatology, Guangzhou Women and Children’s Medical Center, Guangzhou, China
| | - Taoyuan Huang
- Department of Dermatology, Dermatology Hospital of Southern Medical University, Guangzhou, China
| | - Xiaoli Li
- Department of Dermatology, Guangzhou Women and Children’s Medical Center, Guangzhou, China
| | - Peiyi Luo
- Department of Dermatology, Guangzhou Women and Children’s Medical Center, Guangzhou, China
| | - Bingjun Zhang
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
16
|
Li J, Wang Z, Dong H, Hao Y, Gao P, Li W. Different expression levels of interleukin-36 in asthma phenotypes. ALLERGY, ASTHMA, AND CLINICAL IMMUNOLOGY : OFFICIAL JOURNAL OF THE CANADIAN SOCIETY OF ALLERGY AND CLINICAL IMMUNOLOGY 2024; 20:3. [PMID: 38218943 PMCID: PMC10787970 DOI: 10.1186/s13223-023-00868-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Accepted: 06/26/2023] [Indexed: 01/15/2024]
Abstract
Interleukin (IL)-36 family is closely associated with inflammation and consists of IL-36α, IL-36β, IL-36γ, and IL-36Ra. The role of IL-36 in the context of asthma and asthmatic phenotypes is not well characterized. We examined the sputum IL-36 levels in patients with different asthma phenotypes in order to unravel the mechanism of IL-36 in different asthma phenotypes. Our objective was to investigate the induced sputum IL-36α, IL-36β, IL-36γ, and IL-36Ra concentrations in patients with mild asthma, and to analyze the relationship of these markers with lung function and other cytokines in patients with different asthma phenotypes. Induced sputum samples were collected from patients with mild controlled asthma (n = 62, 27 males, age 54.77 ± 15.49) and healthy non-asthmatic controls (n = 16, 10 males, age 54.25 ± 14.60). Inflammatory cell counts in sputum were determined. The concentrations of IL-36 and other cytokines in the sputum supernatant were measured by ELISA and Cytometric Bead Array. This is the first study to report the differential expression of different isoforms of IL-36 in different asthma phenotypes. IL-36α and IL-36β concentrations were significantly higher in the asthma group (P = 0.003 and 0.031), while IL-36Ra concentrations were significantly lower (P < 0.001) compared to healthy non-asthmatic controls. Sputum IL-36α and IL-36β concentrations in the neutrophilic asthma group were significantly higher than those in paucigranulocytic asthma (n = 24) and eosinophilic asthma groups (n = 23). IL-36α and IL-36β showed positive correlation with sputum neutrophils and total cell count (R = 0.689, P < 0.01; R = 0.304, P = 0.008; R = 0.689, P < 0.042; R = 0.253, P = 0.026). In conclusion, IL-36α and IL-36β may contribute to asthma airway inflammation by promoting neutrophil recruitment in airways. Our study provides insights into the inflammatory pathways of neutrophilic asthma and identifies potential therapeutic target.
Collapse
Affiliation(s)
- Jinyan Li
- Department of Respiratory and Critical Care Medicine, The Second Hospital of Jilin University, Changchun, Jilin, China
| | - Zhengda Wang
- Department of Respiratory and Critical Care Medicine, The Second Hospital of Jilin University, Changchun, Jilin, China
| | - Hongna Dong
- Department of Respiratory and Critical Care Medicine, The Second Hospital of Jilin University, Changchun, Jilin, China
| | - Yuqiu Hao
- Department of Respiratory and Critical Care Medicine, The Second Hospital of Jilin University, Changchun, Jilin, China
| | - Peng Gao
- Department of Respiratory and Critical Care Medicine, The Second Hospital of Jilin University, Changchun, Jilin, China.
| | - Wei Li
- Department of Respiratory and Critical Care Medicine, The Second Hospital of Jilin University, Changchun, Jilin, China.
| |
Collapse
|
17
|
Deng L, Costa F, Blake KJ, Choi S, Chandrabalan A, Yousuf MS, Shiers S, Dubreuil D, Vega-Mendoza D, Rolland C, Deraison C, Voisin T, Bagood MD, Wesemann L, Frey AM, Palumbo JS, Wainger BJ, Gallo RL, Leyva-Castillo JM, Vergnolle N, Price TJ, Ramachandran R, Horswill AR, Chiu IM. S. aureus drives itch and scratch-induced skin damage through a V8 protease-PAR1 axis. Cell 2023; 186:5375-5393.e25. [PMID: 37995657 PMCID: PMC10669764 DOI: 10.1016/j.cell.2023.10.019] [Citation(s) in RCA: 38] [Impact Index Per Article: 38.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 08/20/2023] [Accepted: 10/18/2023] [Indexed: 11/25/2023]
Abstract
Itch is an unpleasant sensation that evokes a desire to scratch. The skin barrier is constantly exposed to microbes and their products. However, the role of microbes in itch generation is unknown. Here, we show that Staphylococcus aureus, a bacterial pathogen associated with itchy skin diseases, directly activates pruriceptor sensory neurons to drive itch. Epicutaneous S. aureus exposure causes robust itch and scratch-induced damage. By testing multiple isogenic bacterial mutants for virulence factors, we identify the S. aureus serine protease V8 as a critical mediator in evoking spontaneous itch and alloknesis. V8 cleaves proteinase-activated receptor 1 (PAR1) on mouse and human sensory neurons. Targeting PAR1 through genetic deficiency, small interfering RNA (siRNA) knockdown, or pharmacological blockade decreases itch and skin damage caused by V8 and S. aureus exposure. Thus, we identify a mechanism of action for a pruritogenic bacterial factor and demonstrate the potential of inhibiting V8-PAR1 signaling to treat itch.
Collapse
Affiliation(s)
- Liwen Deng
- Department of Immunology, Harvard Medical School, Boston, MA 02215, USA
| | - Flavia Costa
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Kimbria J Blake
- Department of Immunology, Harvard Medical School, Boston, MA 02215, USA
| | - Samantha Choi
- Department of Immunology, Harvard Medical School, Boston, MA 02215, USA
| | - Arundhasa Chandrabalan
- Department of Physiology and Pharmacology, University of Western Ontario, London, Ontario N6A 5C1, Canada
| | - Muhammad Saad Yousuf
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, TX 75080, USA
| | - Stephanie Shiers
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, TX 75080, USA
| | - Daniel Dubreuil
- Departments of Neurology and Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Daniela Vega-Mendoza
- Division of Immunology, Boston Children's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Corinne Rolland
- IRSD, Université de Toulouse, INSERM, INRAe, ENVT, Université Toulouse III-Paul Sabatier (UPS), Toulouse, France
| | - Celine Deraison
- IRSD, Université de Toulouse, INSERM, INRAe, ENVT, Université Toulouse III-Paul Sabatier (UPS), Toulouse, France
| | - Tiphaine Voisin
- Department of Immunology, Harvard Medical School, Boston, MA 02215, USA
| | - Michelle D Bagood
- Department of Dermatology, University of California, San Diego, La Jolla, CA 92093, USA
| | - Lucia Wesemann
- Department of Immunology, Harvard Medical School, Boston, MA 02215, USA
| | - Abigail M Frey
- Department of Immunology, Harvard Medical School, Boston, MA 02215, USA
| | - Joseph S Palumbo
- Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center and the University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Brian J Wainger
- Departments of Neurology and Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Richard L Gallo
- Department of Dermatology, University of California, San Diego, La Jolla, CA 92093, USA
| | | | - Nathalie Vergnolle
- IRSD, Université de Toulouse, INSERM, INRAe, ENVT, Université Toulouse III-Paul Sabatier (UPS), Toulouse, France
| | - Theodore J Price
- Department of Neuroscience and Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, TX 75080, USA
| | - Rithwik Ramachandran
- Department of Physiology and Pharmacology, University of Western Ontario, London, Ontario N6A 5C1, Canada
| | - Alexander R Horswill
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Isaac M Chiu
- Department of Immunology, Harvard Medical School, Boston, MA 02215, USA.
| |
Collapse
|
18
|
Wang L, Tu Y, Wu W, Tu Y, Yang Z, Chai Y, Yang X, He L. Role of interleukin-36γ induced by ultraviolet radiation in chronic actinic dermatitis. PHOTODERMATOLOGY, PHOTOIMMUNOLOGY & PHOTOMEDICINE 2023; 39:598-606. [PMID: 37545106 DOI: 10.1111/phpp.12903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Revised: 06/20/2023] [Accepted: 07/14/2023] [Indexed: 08/08/2023]
Abstract
BACKGROUND Chronic actinic dermatitis (CAD) is an immune-mediated photodermatosis characterized by a high eosinophil count and total immunoglobulin E (IgE) in the peripheral blood of patients. At present, however, the reasons for their elevation remain unclear. OBJECTIVE The current study aimed to detect changes in inflammatory cytokines in CAD and explore their role in this disease. METHODS Enzyme-linked immunosorbent assay and Luminex assay were conducted to measure inflammatory factor levels. Immunohistochemical analysis and quantitative real-time polymerase chain reaction were performed to evaluate the expression levels of interleukin-36γ (IL-36γ), IL-8, chemokine (C-C motif) ligand 17 (CCL17), and CCL18. CCK8 kits were used to assess cell proliferation. Immunofluorescence was used to detect nuclear factor κB (NF-κB) p65 nuclear translocation. Western blot analysis was performed to detect the protein expression level of phosphorylated NF-κB (p-NF-κB) p65. Hematoxylin and eosin and Masson trichrome staining were applied to observe histological changes in a chronic photo-damaged mouse model. RESULTS Eosinophils, total IgE, IL-36γ, IL-8, tumor necrosis factor α, CCL17, and CCL18 were elevated in CAD. Of note, IL-36γ promoted the proliferation of eosinophilic cells (EOL-1) and the production of IgE in peripheral blood mononuclear cells. IL-36γ also promoted the production of IL-8 and CCL18 in immortalized human keratinocytes (HaCaT cells), while ultraviolet radiation (UVR)-induced IL-36γ via activation of the NF-κB signaling pathway. CONCLUSIONS IL-36γ was involved in the pathogenesis of CAD and UVR contributed to the production of IL-36γ, which may provide a novel therapeutic target for CAD.
Collapse
Affiliation(s)
- Li Wang
- Department of Dermatology, Wuhan No. 1 Hospital, Hubei, China
- Department of Dermatology, First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Yunhua Tu
- Department of Dermatology, First Affiliated Hospital of Kunming Medical University, Kunming, China
- Department of Dermatology, Second People's Hospital of Guiyang, Guiyang, China
| | - Wenjuan Wu
- Department of Dermatology, First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Ying Tu
- Department of Dermatology, First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Zhenghui Yang
- Department of Dermatology, First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Yanjie Chai
- Department of Dermatology, First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Xinwang Yang
- Department of Anatomy and Histology & Embryology, Faculty of Basic Medical Science, Kunming Medical University, Kunming, Yunnan, China
| | - Li He
- Department of Dermatology, First Affiliated Hospital of Kunming Medical University, Kunming, China
| |
Collapse
|
19
|
Williams MR, Bagood MD, Enroth TJ, Bunch ZL, Jiang N, Liu E, Almoughrabie S, Khalil S, Li F, Brinton S, Cech NB, Horswill AR, Gallo RL. Staphylococcus epidermidis activates keratinocyte cytokine expression and promotes skin inflammation through the production of phenol-soluble modulins. Cell Rep 2023; 42:113024. [PMID: 37610872 PMCID: PMC10586132 DOI: 10.1016/j.celrep.2023.113024] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 06/05/2023] [Accepted: 08/10/2023] [Indexed: 08/25/2023] Open
Abstract
Staphylococcus epidermidis is a common microbe on human skin and has beneficial functions in the skin microbiome. However, under conditions of allergic inflammation, the abundance of S. epidermidis increases, establishing potential danger to the epidermis. To understand how this commensal may injure the host, we investigate phenol-soluble modulin (PSM) peptides produced by S. epidermidis that are similar to peptides produced by Staphylococcus aureus. Synthetic S. epidermidis PSMs induce expression of host defense genes and are cytotoxic to human keratinocytes. Deletion mutants of S. epidermidis lacking these gene products support these observations and further show that PSMs require the action of the EcpA bacterial protease to induce inflammation when applied on mouse skin with an intact stratum corneum. The expression of PSMδ from S. epidermidis is also found to correlate with disease severity in patients with atopic dermatitis. These observations show how S. epidermidis PSMs can promote skin inflammation.
Collapse
Affiliation(s)
- Michael R Williams
- Department of Dermatology, University of California, San Diego, San Diego, CA 92093, USA
| | - Michelle D Bagood
- Department of Dermatology, University of California, San Diego, San Diego, CA 92093, USA
| | - Timothy J Enroth
- Department of Veterans Affairs Denver Health Care System, Denver, CO, USA; Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Zoie L Bunch
- Department of Chemistry and Biochemistry, University of North Carolina Greensboro, Greensboro, NC 27402, USA
| | - Nina Jiang
- Department of Dermatology, University of California, San Diego, San Diego, CA 92093, USA
| | - Edward Liu
- Department of Dermatology, University of California, San Diego, San Diego, CA 92093, USA
| | - Samia Almoughrabie
- Department of Dermatology, University of California, San Diego, San Diego, CA 92093, USA
| | - Shadi Khalil
- Department of Dermatology, University of California, San Diego, San Diego, CA 92093, USA
| | - Fengwu Li
- Department of Dermatology, University of California, San Diego, San Diego, CA 92093, USA
| | - Samantha Brinton
- Department of Dermatology, University of California, San Diego, San Diego, CA 92093, USA
| | - Nadja B Cech
- Department of Chemistry and Biochemistry, University of North Carolina Greensboro, Greensboro, NC 27402, USA
| | - Alexander R Horswill
- Department of Veterans Affairs Denver Health Care System, Denver, CO, USA; Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Richard L Gallo
- Department of Dermatology, University of California, San Diego, San Diego, CA 92093, USA.
| |
Collapse
|
20
|
Gu J, Qin G, Jiang L, Xu W, Wang Y, Liao J, Pan H, Liang Z. Correlations between IL-36 family cytokines in peripheral blood and subjective and objective assessment results in patients with allergic rhinitis. ALLERGY, ASTHMA, AND CLINICAL IMMUNOLOGY : OFFICIAL JOURNAL OF THE CANADIAN SOCIETY OF ALLERGY AND CLINICAL IMMUNOLOGY 2023; 19:79. [PMID: 37649097 PMCID: PMC10470177 DOI: 10.1186/s13223-023-00834-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Accepted: 08/14/2023] [Indexed: 09/01/2023]
Abstract
BACKGROUND Interleukin (IL)-36 family cytokines have received increasing attention, especially in the fields of inflammation and immunity research. However, whether IL-36 family cytokine levels are correlated with the results of the assessment of allergic rhinitis (AR) and affect the severity of AR remains unknown. Therefore, this study aimed to investigate the correlations between IL-36 family cytokine levels and subjective and objective assessment results and to further analyze the possible mechanisms of IL-36 family cytokines in the development of AR. METHODS An enzyme-linked immunosorbent assay (ELISA) was used to detect the concentrations of the IL-36 family cytokines IL-36α, IL-36β, IL-36γ, IL-36Ra, and IL-38 in the peripheral blood of patients with AR. The condition of patients with AR was assessed by 22-item sino-nasal outcome test (SNOT-22) score, visual analogue scale (VAS) scores for disease severity, and serum inhalant allergen immunoglobulin E (IgE) detection. Correlations between IL-36 family cytokine levels and subjective and objective assessment results in patients with AR were analyzed. RESULTS The concentration of IL-36α in the peripheral blood of patients with AR was the highest, and the concentration of IL-36β was the lowest. The concentration of IL-36α was higher in juvenile patients than in adult patients, and there was a difference in the IL-36Ra level between the perennial allergen group and the seasonal allergen group. There was a positive correlation between IL-36α level and IL-36γ level, IL-36γ level and IL-36Ra level, and IL-36Ra level and IL-38 level, and IL-36β level was positively correlated with IL-36Ra and IL-38 levels, respectively. IL-36α level was positively correlated with VAS score for nasal congestion symptom. IL-36β level was positively correlated with the total VAS score for ocular symptoms and VAS scores for ocular itching and eye pain symptoms. However, there was no correlation between the levels of all cytokines in IL-36 family and SNOT-22 score, the number of positive inhaled allergens, or the highest positive intensity of allergen specific immunoglobulin E (sIgE). CONCLUSION Peripheral blood IL-36 family cytokines play an important role in AR, and the concentrations of IL-36α and IL-36β were related to the severity of symptoms in patients with AR.
Collapse
Affiliation(s)
- Jia Gu
- Department of Otolaryngology Head and Neck Surgery, The Affiliated Hospital of Southwest Medical University, Taiping Street & NO. 25, Luzhou, 646000, Sichuan, China
| | - Gang Qin
- Department of Otolaryngology Head and Neck Surgery, The Affiliated Hospital of Southwest Medical University, Taiping Street & NO. 25, Luzhou, 646000, Sichuan, China
| | - Liang Jiang
- Department of Otolaryngology Head and Neck Surgery, The Affiliated Hospital of Southwest Medical University, Taiping Street & NO. 25, Luzhou, 646000, Sichuan, China
| | - Wei Xu
- Department of Otolaryngology Head and Neck Surgery, The Affiliated Hospital of Southwest Medical University, Taiping Street & NO. 25, Luzhou, 646000, Sichuan, China
| | - Yuanyuan Wang
- Department of Otolaryngology Head and Neck Surgery, The Affiliated Hospital of Southwest Medical University, Taiping Street & NO. 25, Luzhou, 646000, Sichuan, China
| | - Jiangxue Liao
- Department of Otolaryngology Head and Neck Surgery, The Affiliated Hospital of Southwest Medical University, Taiping Street & NO. 25, Luzhou, 646000, Sichuan, China
| | - Hongzhu Pan
- Department of Otolaryngology Head and Neck Surgery, The Affiliated Hospital of Southwest Medical University, Taiping Street & NO. 25, Luzhou, 646000, Sichuan, China
| | - Zhuoping Liang
- Department of Otolaryngology Head and Neck Surgery, The Affiliated Hospital of Southwest Medical University, Taiping Street & NO. 25, Luzhou, 646000, Sichuan, China.
| |
Collapse
|
21
|
Zhao H, Ma X, Song J, Jiang J, Fei X, Luo Y, Ru Y, Luo Y, Gao C, Kuai L, Li B. From gut to skin: exploring the potential of natural products targeting microorganisms for atopic dermatitis treatment. Food Funct 2023; 14:7825-7852. [PMID: 37599562 DOI: 10.1039/d3fo02455e] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/22/2023]
Abstract
Atopic dermatitis (AD) is the most common chronic inflammatory skin disease. Recent studies have revealed that interactions between pathogenic microorganisms, which have a tendency to parasitize the skin of AD patients, play a significant role in the progression of the disease. Furthermore, specific species of commensal bacteria in the human intestinal tract can have a profound impact on the immune system by promoting inflammation and pruritogenesis in AD, while also regulating adaptive immunity. Natural products (NPs) have emerged as promising agents for the treatment of various diseases. Consequently, there is growing interest in utilizing natural products as a novel therapeutic approach for managing AD, with a focus on modulating both skin and gut microbiota. In this review, we discuss the mechanisms and interplay between the skin and gut microbiota in relation to AD. Additionally, we provide a comprehensive overview of recent clinical and fundamental research on NPs targeting the skin and gut microbiota for AD treatment. We anticipate that our work will contribute to the future development of NPs and facilitate research on microbial mechanisms, based on the efficacy of NPs in treating AD.
Collapse
Affiliation(s)
- Hang Zhao
- Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200437, China
- Institute of Dermatology, Shanghai Academy of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Xin Ma
- Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200437, China
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, 200443, China
| | - Jiankun Song
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, 200443, China
| | - Jingsi Jiang
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, 200443, China
| | - Xiaoya Fei
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, 200443, China
| | - Yue Luo
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, 200443, China
| | - Yi Ru
- Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200437, China
- Institute of Dermatology, Shanghai Academy of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Ying Luo
- Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200437, China
- Institute of Dermatology, Shanghai Academy of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Chunjie Gao
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, 200443, China
| | - Le Kuai
- Department of Dermatology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200437, China
- Institute of Dermatology, Shanghai Academy of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Bin Li
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, 200443, China
- Institute of Dermatology, Shanghai Academy of Traditional Chinese Medicine, Shanghai, 201203, China
| |
Collapse
|
22
|
Gatmaitan JG, Lee JH. Challenges and Future Trends in Atopic Dermatitis. Int J Mol Sci 2023; 24:11380. [PMID: 37511138 PMCID: PMC10380015 DOI: 10.3390/ijms241411380] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 07/06/2023] [Accepted: 07/10/2023] [Indexed: 07/30/2023] Open
Abstract
Atopic dermatitis represents a complex and multidimensional interaction that represents potential fields of preventive and therapeutic management. In addition to the treatment armamentarium available for atopic dermatitis, novel drugs targeting significant molecular pathways in atopic dermatitis biologics and small molecules are also being developed given the condition's complex pathophysiology. While most of the patients are expecting better efficacy and long-term control, the response to these drugs would still depend on numerous factors such as complex genotype, diverse environmental triggers and microbiome-derived signals, and, most importantly, dynamic immune responses. This review article highlights the challenges and the recently developed pharmacological agents in atopic dermatitis based on the molecular pathogenesis of this condition, creating a specific therapeutic approach toward a more personalized medicine.
Collapse
Affiliation(s)
- Julius Garcia Gatmaitan
- Department of Dermatology, Seoul St. Mary's Hospital, The Catholic University of Korea, 222, Banpo-daero, Seocho-gu, Seoul 06591, Republic of Korea
- Gatmaitan Medical and Skin Center, Baliuag 3006, Bulacan, Philippines
- Skines Aesthetic and Laser Center, Quezon City 1104, Metro Manila, Philippines
| | - Ji Hyun Lee
- Department of Dermatology, Seoul St. Mary's Hospital, The Catholic University of Korea, 222, Banpo-daero, Seocho-gu, Seoul 06591, Republic of Korea
| |
Collapse
|
23
|
Yamada H, Kaitani A, Izawa K, Ando T, Kamei A, Uchida S, Maehara A, Kojima M, Yamamoto R, Wang H, Nagamine M, Maeda K, Uchida K, Nakano N, Ohtsuka Y, Ogawa H, Okumura K, Shimizu T, Kitaura J. Staphylococcus aureus δ-toxin present on skin promotes the development of food allergy in a murine model. Front Immunol 2023; 14:1173069. [PMID: 37275864 PMCID: PMC10235538 DOI: 10.3389/fimmu.2023.1173069] [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: 02/24/2023] [Accepted: 05/05/2023] [Indexed: 06/07/2023] Open
Abstract
Background Patients with food allergy often suffer from atopic dermatitis, in which Staphylococcus aureus colonization is frequently observed. Staphylococcus aureus δ-toxin activates mast cells and promotes T helper 2 type skin inflammation in the tape-stripped murine skin. However, the physiological effects of δ-toxin present on the steady-state skin remain unknown. We aimed to investigate whether δ-toxin present on the steady-state skin impacts the development of food allergy. Material and methods The non-tape-stripped skins of wild-type, KitW-sh/W-sh, or ST2-deficient mice were treated with ovalbumin (OVA) with or without δ-toxin before intragastric administration of OVA. The frequency of diarrhea, numbers of jejunum or skin mast cells, and serum levels of OVA-specific IgE were measured. Conventional dendritic cell 2 (cDC2) in skin and lymph nodes (LN) were analyzed. The cytokine levels in the skin tissues or culture supernatants of δ-toxin-stimulated murine keratinocytes were measured. Anti-IL-1α antibody-pretreated mice were analyzed. Results Stimulation with δ-toxin induced the release of IL-1α, but not IL-33, in murine keratinocytes. Epicutaneous treatment with OVA and δ-toxin induced the local production of IL-1α. This treatment induced the translocation of OVA-loaded cDC2 from skin to draining LN and OVA-specific IgE production, independently of mast cells and ST2. This resulted in OVA-administered food allergic responses. In these models, pretreatment with anti-IL-1α antibody inhibited the cDC2 activation and OVA-specific IgE production, thereby dampening food allergic responses. Conclusion Even without tape stripping, δ-toxin present on skin enhances epicutaneous sensitization to food allergen in an IL-1α-dependent manner, thereby promoting the development of food allergy.
Collapse
Affiliation(s)
- Hiromichi Yamada
- Atopy (Allergy) Research Center, Juntendo University Graduate School of Medicine, Tokyo, Japan
- Department of Pediatrics and Adolescent Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Ayako Kaitani
- Atopy (Allergy) Research Center, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Kumi Izawa
- Atopy (Allergy) Research Center, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Tomoaki Ando
- Atopy (Allergy) Research Center, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Anna Kamei
- Atopy (Allergy) Research Center, Juntendo University Graduate School of Medicine, Tokyo, Japan
- Department of Science of Allergy and Inflammation, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Shino Uchida
- Atopy (Allergy) Research Center, Juntendo University Graduate School of Medicine, Tokyo, Japan
- Department of Gastroenterology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Akie Maehara
- Atopy (Allergy) Research Center, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Mayuki Kojima
- Atopy (Allergy) Research Center, Juntendo University Graduate School of Medicine, Tokyo, Japan
- Department of Pediatrics and Adolescent Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Risa Yamamoto
- Atopy (Allergy) Research Center, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Hexing Wang
- Atopy (Allergy) Research Center, Juntendo University Graduate School of Medicine, Tokyo, Japan
- Department of Science of Allergy and Inflammation, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Masakazu Nagamine
- Atopy (Allergy) Research Center, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Keiko Maeda
- Atopy (Allergy) Research Center, Juntendo University Graduate School of Medicine, Tokyo, Japan
- Department of Immunological Diagnosis, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Koichiro Uchida
- Atopy (Allergy) Research Center, Juntendo University Graduate School of Medicine, Tokyo, Japan
- Juntendo Advanced Research Institute for Health Science, Juntendo University School of Medicine, Tokyo, Japan
| | - Nobuhiro Nakano
- Atopy (Allergy) Research Center, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Yoshikazu Ohtsuka
- Department of Pediatrics and Adolescent Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Hideoki Ogawa
- Atopy (Allergy) Research Center, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Ko Okumura
- Atopy (Allergy) Research Center, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Toshiaki Shimizu
- Atopy (Allergy) Research Center, Juntendo University Graduate School of Medicine, Tokyo, Japan
- Department of Pediatrics and Adolescent Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Jiro Kitaura
- Atopy (Allergy) Research Center, Juntendo University Graduate School of Medicine, Tokyo, Japan
- Department of Science of Allergy and Inflammation, Juntendo University Graduate School of Medicine, Tokyo, Japan
| |
Collapse
|
24
|
Schuler CF, Billi AC, Maverakis E, Tsoi LC, Gudjonsson JE. Novel insights into atopic dermatitis. J Allergy Clin Immunol 2023; 151:1145-1154. [PMID: 36428114 PMCID: PMC10164702 DOI: 10.1016/j.jaci.2022.10.023] [Citation(s) in RCA: 48] [Impact Index Per Article: 48.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 10/20/2022] [Accepted: 10/24/2022] [Indexed: 11/23/2022]
Abstract
Recent research into the pathophysiology and treatment of atopic dermatitis (AD) has shown notable progress. An increasing number of aspects of the immune system are being implicated in AD, including the epithelial barrier, TH2 cytokines, and mast cells. Major advances in therapeutics were made in biologic cytokine and receptor antagonists and among Janus kinase inhibitors. We focus on these areas and address new insights into AD epidemiology, biomarkers, endotypes, prevention, and comorbidities. Going forward, we expect future mechanistic insights and therapeutic advances to broaden physicians' ability to diagnose and manage AD patients, and perhaps to find a cure for this chronic condition.
Collapse
Affiliation(s)
- Charles F Schuler
- Division of Allergy and Clinical Immunology, Department of Internal Medicine, Michigan Medicine, University of Michigan, Ann Arbor, Mich; Mary H. Weiser Food Allergy Center, University of Michigan, Ann Arbor, Mich
| | - Allison C Billi
- Department of Dermatology, Michigan Medicine, University of Michigan, Ann Arbor, Mich
| | - Emanual Maverakis
- Department of Dermatology, University of California-Davis, Sacramento, Calif
| | - Lam C Tsoi
- Department of Dermatology, Michigan Medicine, University of Michigan, Ann Arbor, Mich
| | - Johann E Gudjonsson
- Department of Dermatology, Michigan Medicine, University of Michigan, Ann Arbor, Mich.
| |
Collapse
|
25
|
Rauer L, Reiger M, Bhattacharyya M, Brunner PM, Krueger JG, Guttman-Yassky E, Traidl-Hoffmann C, Neumann AU. Skin microbiome and its association with host cofactors in determining atopic dermatitis severity. J Eur Acad Dermatol Venereol 2023; 37:772-782. [PMID: 36433676 DOI: 10.1111/jdv.18776] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Accepted: 11/09/2022] [Indexed: 11/27/2022]
Abstract
BACKGROUND Atopic dermatitis (AD) is a heterogeneous, chronic inflammatory skin disease linked to skin microbiome dysbiosis with reduced bacterial diversity and elevated relative abundance of Staphylococcus aureus (S. aureus). OBJECTIVES We aimed to characterize the yet incompletely understood association between the skin microbiome and patients' demographic and clinical cofactors in relation to AD severity. METHODS The skin microbiome in 48 adult moderate-to-severe AD patients was investigated using next-generation deep sequencing (16S rRNA gene, V1-V3 region) followed by denoising (DADA2) to obtain amplicon sequence variant (ASV) composition. RESULTS In lesional skin, AD severity was associated with S. aureus relative abundance (rS = 0.53, p < 0.001) and slightly better with the microbiome diversity measure Evenness (rS = -0.58, p < 0.001), but not with Richness. Multiple regression confirmed the association of AD severity with microbiome diversity, including Shannon (in lesional skin, p < 0.001), Evenness (in non-lesional skin, p = 0.015) or S. aureus relative abundance (p < 0.012), and with patient's IgE levels (p < 0.001), race (p < 0.032), age (p < 0.034) and sex (p = 0.012). The lesional model explained 62% of the variation in AD severity, and the non-lesional model 50% of the variation. CONCLUSIONS Our results specify the frequently reported "reduced diversity" of the AD-related skin microbiome to reduced Evenness, which was in turn mainly driven by S. aureus relative abundance, rather than to a reduced microbiome Richness. Finding associations between AD severity, the skin microbiome and patient's cofactors is a key aspect in developing new personalized AD treatments, particularly those targeting the AD microbiome.
Collapse
Affiliation(s)
- Luise Rauer
- Environmental Medicine, Faculty of Medicine, University of Augsburg, Augsburg, Germany.,Chair of Environmental Medicine, Technical University Munich, Munich, Germany.,Institute of Environmental Medicine, Helmholtz Zentrum München, Augsburg, Germany.,Institute for Medical Information Processing, Biometry and Epidemiology (IBE), LMU, Munich, Germany
| | - Matthias Reiger
- Environmental Medicine, Faculty of Medicine, University of Augsburg, Augsburg, Germany.,Chair of Environmental Medicine, Technical University Munich, Munich, Germany.,Institute of Environmental Medicine, Helmholtz Zentrum München, Augsburg, Germany
| | - Madhumita Bhattacharyya
- Environmental Medicine, Faculty of Medicine, University of Augsburg, Augsburg, Germany.,Chair of Environmental Medicine, Technical University Munich, Munich, Germany
| | - Patrick M Brunner
- Laboratory for Investigative Dermatology, The Rockefeller University, New York, New York, USA.,Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - James G Krueger
- Laboratory for Investigative Dermatology, The Rockefeller University, New York, New York, USA
| | - Emma Guttman-Yassky
- Laboratory for Investigative Dermatology, The Rockefeller University, New York, New York, USA.,Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Claudia Traidl-Hoffmann
- Environmental Medicine, Faculty of Medicine, University of Augsburg, Augsburg, Germany.,Chair of Environmental Medicine, Technical University Munich, Munich, Germany.,Institute of Environmental Medicine, Helmholtz Zentrum München, Augsburg, Germany.,CK-CARE Center for Allergy Research and Education, Davos, Switzerland.,ZIEL - Institute for Food & Health, Technical University of Munich, Freising, Germany
| | - Avidan U Neumann
- Environmental Medicine, Faculty of Medicine, University of Augsburg, Augsburg, Germany.,Institute of Environmental Medicine, Helmholtz Zentrum München, Augsburg, Germany.,CK-CARE Center for Allergy Research and Education, Davos, Switzerland
| |
Collapse
|
26
|
Youn C, Dikeman DA, Chang E, Liu H, Nolan SJ, Alphonse MP, Joyce DP, Liu Q, Meixiong J, Dong X, Miller LS, Archer NK. Crisaborole efficacy in murine models of skin inflammation and Staphylococcus aureus infection. Exp Dermatol 2023; 32:425-435. [PMID: 36461082 PMCID: PMC10066830 DOI: 10.1111/exd.14722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 09/09/2022] [Accepted: 11/28/2022] [Indexed: 12/05/2022]
Abstract
Phosphodiesterase 4 (PDE4) is highly expressed in keratinocytes and immune cells and promotes pro-inflammatory responses upon activation. The activity of PDE4 has been attributed to various inflammatory conditions, leading to the development and approval of PDE4 inhibitors as host-directed therapeutics in humans. For example, the topical PDE4 inhibitor, crisaborole, is approved for the treatment of mild-to-moderate atopic dermatitis and has shown efficacy in patients with psoriasis. However, the role of crisaborole in regulating the immunopathogenesis of inflammatory skin diseases and infection is not entirely known. Therefore, we evaluated the effects of crisaborole in multiple mouse models, including psoriasis-like dermatitis, AD-like skin inflammation with and without filaggrin mutations, and Staphylococcus aureus skin infection. We discovered that crisaborole dampens myeloid cells and itch in the skin during psoriasis-like dermatitis. Furthermore, crisaborole was effective in reducing skin inflammation in the context of filaggrin deficiency. Importantly, crisaborole reduced S. aureus skin colonization during AD-like skin inflammation. However, crisaborole was not efficacious in treating S. aureus skin infections, even as adjunctive therapy to antibiotics. Taken together, we found that crisaborole reduced itch during psoriasis-like dermatitis and decreased S. aureus skin colonization upon AD-like skin inflammation, which act as additional mechanisms by which crisaborole dampens the immunopathogenesis in mouse models of inflammatory skin diseases. Further examination is warranted to translate these preclinical findings to human disease.
Collapse
Affiliation(s)
- Christine Youn
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Dustin A. Dikeman
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Evelyn Chang
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Haiyun Liu
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Sabrina J. Nolan
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Martin P. Alphonse
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Daniel P. Joyce
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Qi Liu
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - James Meixiong
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Xinzhong Dong
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Howard Hughes Medical Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Lloyd S. Miller
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Nathan K. Archer
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
27
|
Maglie R, Mercurio L, Morelli M, Madonna S, Salemme A, Baffa ME, Quintarelli L, Di Zenzo GM, Antiga E, Albanesi C. Interleukin-36 cytokines are overexpressed in the skin and sera of patients with bullous pemphigoid. Exp Dermatol 2023. [PMID: 36940975 DOI: 10.1111/exd.14791] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 02/21/2023] [Accepted: 02/28/2023] [Indexed: 03/22/2023]
Abstract
Bullous pemphigoid (BP) is an autoimmune bullous disease, characterized by autoantibodies targeting BP180 and BP230. The role of interleukin (IL)-36, a potent chemoattractant for granulocytes, in BP remains elusive.The expression of IL-36 cytokines (IL-36α, β, γ) and their antagonists (IL-36Ra and IL-38) was analysed in the skin and serum samples of patients with BP (n = 31), psoriasis (n = 10) and healthy controls (HC) (n = 14) by quantitative polymerase chain reaction and enzyme linked immunosorbent assay, respectively. Skin and serum levels of all cytokines were correlated with the Bullous Pemphigoid Disease Area Index (BPDAI) score and with the serum concentration of pathogenic antibodies.IL-36α, IL-36β, IL-36γ and IL-36Ra were significantly (p < 0.05) overexpressed in BP skin compared to HC, without remarkable differences relative to psoriasis skin. The expression of IL-38 was significantly (p < 0.05) higher in BP compared to psoriasis skin.IL-36α and γ, but not β, serum concentrations were significantly (p < 0.05) higher in BP compared to HC. IL-36γ was significantly (p < 0.05) more expressed in the serum of psoriasis patients than BP. The serum concentration of IL-36Ra and IL-38 were similar between BP and HC, while IL-38 serum levels were significantly (p < 0.05) higher in BP compared to psoriasis patients. Serum IL-36α correlated significantly with BPDAI (r = 0.5 p = 0.001).IL-36 agonists are increased in BP patients, both locally and systemically. Serum IL-36α might represent a potential biomarker for BP. An inefficient balance between IL-36 agonists and antagonists is likely to occur during BP inflammation.
Collapse
Affiliation(s)
- Roberto Maglie
- Section of Dermatology, Department of Health Sciences, University of Florence, Florence, Italy
| | - Laura Mercurio
- Experimental Immunology Laboratory, Istituto Dermopatico dell'Immacolata (IDI-IRCCS), Rome, Italy
| | - Martina Morelli
- Experimental Immunology Laboratory, Istituto Dermopatico dell'Immacolata (IDI-IRCCS), Rome, Italy
| | - Stefania Madonna
- Experimental Immunology Laboratory, Istituto Dermopatico dell'Immacolata (IDI-IRCCS), Rome, Italy
| | - Adele Salemme
- Molecular and Cell Biology laboratory, Istituto Dermopatico dell'Immacolata (IDI-IRCCS), Rome, Italy
| | - Maria E Baffa
- Section of Dermatology, Department of Health Sciences, University of Florence, Florence, Italy
| | - Lavinia Quintarelli
- Section of Dermatology, Department of Health Sciences, University of Florence, Florence, Italy
| | - Giovanni M Di Zenzo
- Molecular and Cell Biology laboratory, Istituto Dermopatico dell'Immacolata (IDI-IRCCS), Rome, Italy
| | - Emiliano Antiga
- Section of Dermatology, Department of Health Sciences, University of Florence, Florence, Italy
| | - Cristina Albanesi
- Experimental Immunology Laboratory, Istituto Dermopatico dell'Immacolata (IDI-IRCCS), Rome, Italy
| |
Collapse
|
28
|
Abstract
Interleukin (IL)-36 cytokines are members of the IL-1 superfamily of cytokines. IL-36 cytokines are composed of three agonists (IL-36α, IL-36β, and IL-36γ) and two antagonists (IL-36 receptor antagonist [IL36Ra] and IL-38). These work in innate and acquired immunity and are known to contribute to host defense and to the pathogenesis of autoinflammatory diseases, autoimmune diseases, and infectious diseases. In the skin, IL-36α and IL-36γ are mainly expressed by keratinocytes in the epidermis, although they are also produced by dendritic cells, macrophages, endothelial cells, and dermal fibroblasts. IL-36 cytokines participate in the first-line defense of the skin against various exogenous assaults. IL-36 cytokines play significant roles in the host defense system and in the regulation of inflammatory pathways in the skin, collaborating with other cytokines/chemokines and immune-related molecules. Thus, numerous studies have shown IL-36 cytokines to play important roles in the pathogenesis of various skin diseases. In this context, the clinical efficacy and safety profiles of anti-IL-36 agents such as spesolimab and imsidolimab have been evaluated in patients with generalized pustular psoriasis, palmoplantar pustulosis, hidradenitis suppurativa, acne/acneiform eruptions, ichthyoses, and atopic dermatitis. This article comprehensively summarizes the roles played by IL-36 cytokines in the pathogenesis and pathophysiology of various skin diseases and summarizes the current state of research on therapeutic agents that target IL-36 cytokine pathways.
Collapse
|
29
|
Bissonnette R, Abramovits W, Saint-Cyr Proulx É, Lee P, Guttman-Yassky E, Zovko E, Sigmund R, Willcox J, Bieber T. Spesolimab, an anti-interleukin-36 receptor antibody, in patients with moderate-to-severe atopic dermatitis: Results from a multicentre, randomized, double-blind, placebo-controlled, phase IIa study. J Eur Acad Dermatol Venereol 2023; 37:549-557. [PMID: 36376738 DOI: 10.1111/jdv.18727] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 10/13/2022] [Indexed: 11/17/2022]
Abstract
BACKGROUND Atopic dermatitis (AD) is a chronic relapsing inflammatory skin disease, and there is increasing evidence that the interleukin (IL)-36 pathway may play a role in the pathogenesis of AD. OBJECTIVES To evaluate the efficacy and safety of spesolimab, a novel anti-IL-36 receptor antibody, in adult patients with moderate-to-severe AD. METHODS In this phase IIa study, 51 eligible patients were randomized 2:1 to receive intravenous doses of spesolimab 600 mg or placebo every 4 weeks. The primary endpoint was the percentage change from baseline in Eczema Area and Severity Index (EASI) score at Week 16. RESULTS The decrease in EASI score from baseline to Week 16 was -37.9% for spesolimab versus -12.3% for placebo (adjusted mean difference -25.6%, p = 0.149). A predefined sensitivity analysis, excluding data from patients who used restricted corticosteroids, resulted in an adjusted mean difference of -48.3% (nominal p = 0.024). Spesolimab was well tolerated, with no clinically relevant safety signals. CONCLUSIONS This is the first study to evaluate the IL-36 pathway inhibition in AD. Although not statistically significant, numerical improvements were observed in the primary endpoint of change from baseline in the EASI score. Spesolimab had an acceptable safety profile, with no unexcepted safety concerns.
Collapse
Affiliation(s)
| | | | | | - Patricia Lee
- Center for Clinical Studies, Houston, Texas, USA
| | - Emma Guttman-Yassky
- Department of Dermatology and Immunology Institute, Icahn School of Medicine, Mount Sinai, New York, New York, USA
| | - Elizabeta Zovko
- Boehringer Ingelheim Corporation, Ridgefield, Connecticut, USA
| | - Ralf Sigmund
- Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | - Joanne Willcox
- Boehringer Ingelheim Pharmaceuticals Ltd, Burlington, Ontario, Canada
| | - Thomas Bieber
- Department of Dermatology and Allergy, Christine Kühne-Center for Allergy Research and Education (CK-CARE), University Hospital Bonn, Bonn, Germany
| |
Collapse
|
30
|
Makowska K, Nowaczyk J, Blicharz L, Waśkiel-Burnat A, Czuwara J, Olszewska M, Rudnicka L. Immunopathogenesis of Atopic Dermatitis: Focus on Interleukins as Disease Drivers and Therapeutic Targets for Novel Treatments. Int J Mol Sci 2023; 24:ijms24010781. [PMID: 36614224 PMCID: PMC9820829 DOI: 10.3390/ijms24010781] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 12/20/2022] [Accepted: 12/23/2022] [Indexed: 01/04/2023] Open
Abstract
Atopic dermatitis is a chronic, recurrent inflammatory skin disorder manifesting by eczematous lesions and intense pruritus. Atopic dermatitis develops primarily as a result of an epidermal barrier defect and immunological imbalance. Advances in understanding these pathogenetic hallmarks, and particularly the complex role of interleukins as atopic dermatitis drivers, resulted in achieving significant therapeutic breakthroughs. Novel medications involve monoclonal antibodies specifically blocking the function of selected interleukins and small molecules such as Janus kinase inhibitors limiting downstream signaling to reduce the expression of a wider array of proinflammatory factors. Nevertheless, a subset of patients remains refractory to those treatments, highlighting the complexity of atopic dermatitis immunopathogenesis in different populations. In this review, we address the immunological heterogeneity of atopic dermatitis endotypes and phenotypes and present novel interleukin-oriented therapies for this disease.
Collapse
|
31
|
Lv H, Wang Y, Gao Z, Liu P, Qin D, Hua Q, Xu Y. Knowledge mapping of the links between the microbiota and allergic diseases: A bibliometric analysis (2002-2021). Front Immunol 2022; 13:1045795. [PMID: 36389800 PMCID: PMC9650552 DOI: 10.3389/fimmu.2022.1045795] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Accepted: 10/14/2022] [Indexed: 08/17/2023] Open
Abstract
Background In recent decades, dramatic changes in modern environmental exposures and lifestyles have resulted in a steep rise in the prevalence of allergic diseases such as asthma, allergic rhinitis, atopic dermatitis and food allergies. Evidence is mounting that the microbiota plays a crucial role in allergic disorder development and evolution. Therefore, a better understanding of allergic diseases within the context of the microbiota is urgently needed. This work aimed to comprehensively outline general characteristics, research hotspots, evolution routes, and emerging trends in this area. Methods Relevant publications from January 2002 to December 2021 were obtained from the Web of Science Core Collection on 5 August 2022. Bibliometric and visual analyses were performed using CiteSpace; VOSviewer; an online bibliometric platform; and Microsoft Excel 2019. Results In total, 2535 documents met the requirements. The annual number of publications has shown rapid growth in the last two decades. The USA, University of California System, and Isolauri E of the University of Turku were the most productive and influential country, institution, and author, respectively. The Journal of Allergy and Clinical Immunology was the most prolific and most cocited journal. High-frequency keywords included "gut microbiota", "asthma", "atopic dermatitis", "children", and "probiotics". Recent studies have focused on "atopic dermatitis", "skin", "asthma", and "probiotics", according to the cocitation analysis of references. Burst detection analysis of keywords showed that "community", "skin microbiome", "microbiome", "Staphylococcus aureus", and "chain fatty acid" were emerging research frontiers, which currently have ongoing bursts. Conclusion In the last 20 years, studies of the microbiota in allergic diseases have been flourishing, and the themes have been increasing in depth. These findings provide valuable references on the current research hotspots and gaps and development trends in the link between the microbiota and allergic diseases.
Collapse
Affiliation(s)
- Hao Lv
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yunfei Wang
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Ziang Gao
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Peiqiang Liu
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Danxue Qin
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Qingquan Hua
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan, China
- Research Institute of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yu Xu
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan, China
- Research Institute of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
32
|
Iznardo H, Puig L. IL-1 Family Cytokines in Inflammatory Dermatoses: Pathogenetic Role and Potential Therapeutic Implications. Int J Mol Sci 2022; 23:ijms23169479. [PMID: 36012744 PMCID: PMC9409147 DOI: 10.3390/ijms23169479] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 08/18/2022] [Accepted: 08/19/2022] [Indexed: 12/15/2022] Open
Abstract
The interleukin-1 (IL-1) family is involved in the correct functioning and regulation of the innate immune system, linking innate and adaptative immune responses. This complex family is composed by several cytokines, receptors, and co-receptors, all working in a balanced way to maintain homeostasis. Dysregulation of these processes results in tissue inflammation and is involved in the pathogenesis of common inflammatory dermatoses such as psoriasis, hidradenitis suppurativa, and atopic dermatitis. Therefore, therapeutic targeting of IL-1 pathways has been studied, and several monoclonal antibodies are currently being assessed in clinical trials. So far, promising results have been obtained with anti-IL-36R spesolimab and imsidolimab in pustular psoriasis, and their efficacy is being tested in other conditions.
Collapse
Affiliation(s)
- Helena Iznardo
- Dermatology Department, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, 08041 Barcelona, Spain
- Institut d’Investigació Biomèdica Sant Pau (IIB SANT PAU), 08041 Barcelona, Spain
| | - Luís Puig
- Dermatology Department, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, 08041 Barcelona, Spain
- Institut d’Investigació Biomèdica Sant Pau (IIB SANT PAU), 08041 Barcelona, Spain
- Correspondence:
| |
Collapse
|
33
|
Sachen KL, Arnold Greving CN, Towne JE. Role of IL-36 cytokines in psoriasis and other inflammatory skin conditions. Cytokine 2022; 156:155897. [DOI: 10.1016/j.cyto.2022.155897] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 03/29/2022] [Accepted: 04/22/2022] [Indexed: 12/15/2022]
|
34
|
Merleev A, Ji-Xu A, Toussi A, Tsoi LC, Le ST, Luxardi G, Xing X, Wasikowski R, Liakos W, Brüggen MC, Elder JT, Adamopoulos IE, Izumiya Y, Riera-Leal A, Li Q, Kuzminykh NY, Kirane A, Marusina AI, Gudjonsson JE, Maverakis E. Proprotein convertase subtilisin/kexin type 9 (PCSK9) is a psoriasis susceptibility locus that is negatively related to IL36G. JCI Insight 2022; 7:141193. [PMID: 35862195 PMCID: PMC9462487 DOI: 10.1172/jci.insight.141193] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Accepted: 07/18/2022] [Indexed: 11/17/2022] Open
Abstract
Proprotein convertase subtilisin/kexin type-9 (PCSK9) is a posttranslational regulator of the LDL receptor (LDLR). Recent studies have proposed a role for PCSK9 in regulating immune responses. Using RNA-Seq–based variant discovery, we identified a possible psoriasis-susceptibility locus at 1p32.3, located within PCSK9 (rs662145 C > T). This finding was verified in independently acquired genomic and RNA-Seq data sets. Single-cell RNA-Seq (scRNA-Seq) identified keratinocytes as the primary source of PCSK9 in human skin. PCSK9 expression, however, was not uniform across keratinocyte subpopulations. scRNA-Seq and IHC demonstrated an epidermal gradient of PCSK9, with expression being highest in basal and early spinous layer keratinocytes and lowest in granular layer keratinocytes. IL36G expression followed the opposite pattern, with expression highest in granular layer keratinocytes. PCSK9 siRNA knockdown experiments confirmed this inverse relationship between PCSK9 and IL36G expression. Other immune genes were also linked to PCSK9 expression, including IL27RA, IL1RL1, ISG20, and STX3. In both cultured keratinocytes and nonlesional human skin, homozygosity for PCSK9 SNP rs662145 C > T was associated with lower PCSK9 expression and higher IL36G expression, when compared with heterozygous skin or cell lines. Together, these results support PCSK9 as a psoriasis-susceptibility locus and establish a putative link between PCSK9 and inflammatory cytokine expression.
Collapse
Affiliation(s)
- Alexander Merleev
- Department of Dermatology, University of California at Davis, Sacramento, United States of America
| | - Antonio Ji-Xu
- Department of Dermatology, University of California at Davis, Sacramento, United States of America
| | - Atrin Toussi
- Department of Dermatology, University of California at Davis, Sacramento, United States of America
| | - Lam C Tsoi
- The University of Michigan Medical School, Ann Arbor, United States of America
| | - Stephanie T Le
- Department of Dermatology, University of California at Davis, Sacramento, United States of America
| | - Guillaume Luxardi
- Department of Dermatology, University of California at Davis, Sacramento, United States of America
| | - Xianying Xing
- Department of Dermatology, The University of Michigan Medical School, Ann Arbor, United States of America
| | - Rachael Wasikowski
- Department of Dermatology, The University of Michigan Medical School, Ann Arbor, United States of America
| | - William Liakos
- Department of Dermatology, University of California at Davis, Sacramento, United States of America
| | | | - James T Elder
- The University of Michigan Medical School, Ann Arbor, United States of America
| | - Iannis E Adamopoulos
- Division of Rheumatology and Clinical Immunology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, United States of America
| | - Yoshihiro Izumiya
- Department of Biochemistry and Molecular Medicine, University of California at Davis, Sacramento, United States of America
| | - Annie Riera-Leal
- Department of Dermatology, University of California at Davis, Sacramento, United States of America
| | - Qinyuan Li
- Department of Dermatology, University of California at Davis, Sacramento, United States of America
| | - Nikolay Yu Kuzminykh
- Institute of Biochemical Physics, Russian Academy of Sciences, Moscow, Russian Federation
| | - Amanda Kirane
- Department of Surgical Oncology, University of California at Davis, Sacramento, United States of America
| | - Alina I Marusina
- Department of Dermatology, University of California at Davis, Sacramento, United States of America
| | - Johann E Gudjonsson
- The University of Michigan Medical School, Ann Arbor, United States of America
| | - Emanual Maverakis
- Department of Dermatology, University of California at Davis, Sacramento, United States of America
| |
Collapse
|
35
|
Exploring the Role of Staphylococcus aureus in Inflammatory Diseases. Toxins (Basel) 2022; 14:toxins14070464. [PMID: 35878202 PMCID: PMC9318596 DOI: 10.3390/toxins14070464] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Revised: 06/23/2022] [Accepted: 07/01/2022] [Indexed: 02/04/2023] Open
Abstract
Staphylococcus aureus is a very common Gram-positive bacterium, and S. aureus infections play an extremely important role in a variety of diseases. This paper describes the types of virulence factors involved, the inflammatory cells activated, the process of host cell death, and the associated diseases caused by S. aureus. S. aureus can secrete a variety of enterotoxins and other toxins to trigger inflammatory responses and activate inflammatory cells, such as keratinocytes, helper T cells, innate lymphoid cells, macrophages, dendritic cells, mast cells, neutrophils, eosinophils, and basophils. Activated inflammatory cells can express various cytokines and induce an inflammatory response. S. aureus can also induce host cell death through pyroptosis, apoptosis, necroptosis, autophagy, etc. This article discusses S. aureus and MRSA (methicillin-resistant S. aureus) in atopic dermatitis, psoriasis, pulmonary cystic fibrosis, allergic asthma, food poisoning, sarcoidosis, multiple sclerosis, and osteomyelitis. Summarizing the pathogenic mechanism of Staphylococcus aureus provides a basis for the targeted treatment of Staphylococcus aureus infection.
Collapse
|
36
|
Matarazzo L, Hernandez Santana YE, Walsh PT, Fallon PG. The IL-1 cytokine family as custodians of barrier immunity. Cytokine 2022; 154:155890. [DOI: 10.1016/j.cyto.2022.155890] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 03/31/2022] [Accepted: 04/13/2022] [Indexed: 12/12/2022]
|
37
|
Abstract
Human skin forms a protective barrier against the external environment and is our first line of defense against toxic, solar, and pathogenic insults. Our skin also defines our outward appearance, protects our internal tissues and organs, acts as a sensory interface, and prevents dehydration. Crucial to the skin's barrier function is the colonizing microbiota, which provides protection against pathogens, tunes immune responses, and fortifies the epithelium. Here we highlight recent advances in our understanding of how the microbiota mediates multiple facets of skin barrier function. We discuss recent insights into pathological host-microbiota interactions and implications for disorders of the skin and distant organs. Finally, we examine how microbiota-based mechanisms can be targeted to prevent or manage skin disorders and impaired wound healing.
Collapse
Affiliation(s)
- Tamia A Harris-Tryon
- Department of Dermatology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Elizabeth A Grice
- Departments of Dermatology and Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
38
|
Humeau M, Boniface K, Bodet C. Cytokine-Mediated Crosstalk Between Keratinocytes and T Cells in Atopic Dermatitis. Front Immunol 2022; 13:801579. [PMID: 35464457 PMCID: PMC9022745 DOI: 10.3389/fimmu.2022.801579] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 03/07/2022] [Indexed: 01/22/2023] Open
Abstract
Atopic dermatitis (AD) is a chronic inflammatory skin disease characterized by barrier dysfunction, dysregulated immune response, and dysbiosis with increased Staphylococcus aureus colonization. Infiltration of various T helper cell subsets into lesional skin and subsequent cytokine release are a hallmark of AD. Release of cytokines by both T cells and keratinocytes plays a key role in skin inflammation and drives many AD features. This review aims to discuss cytokine-mediated crosstalk between T cells and keratinocytes in AD pathogenesis and the potential impact of virulence factors produced by Staphylococcus aureus on these interactions.
Collapse
Affiliation(s)
- Mélanie Humeau
- Laboratoire Inflammation Tissus Epithéliaux et Cytokines LITEC UR 15560, Université de Poitiers, Poitiers, France
| | - Katia Boniface
- ImmunoConcEpT, Centre National de la Recherche Scientifique, Unité Mixte de Recherche 5164, University of Bordeaux, Bordeaux, France
| | - Charles Bodet
- Laboratoire Inflammation Tissus Epithéliaux et Cytokines LITEC UR 15560, Université de Poitiers, Poitiers, France
| |
Collapse
|
39
|
Hwang J, Rick J, Hsiao J, Shi VY. A review of IL-36: an emerging therapeutic target for inflammatory dermatoses. J DERMATOL TREAT 2022; 33:2711-2722. [PMID: 35470744 DOI: 10.1080/09546634.2022.2067819] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
BACKGROUND IL-36 cytokines are members of the IL-1 superfamily. Increasing evidence in the IL-36 pathway demonstrates their potential as a therapeutic target for treating inflammatory skin diseases, such as generalized pustular psoriasis (GPP). OBJECTIVE A narrative review was written to further study preclinical and clinical evidence for the role of IL-36 in psoriasis, atopic dermatitis (AD), hidradenitis suppurativa (HS), acne, autoimmune blistering diseases, and neutrophilic dermatoses. RESULTS IL-36 has important downstream effects such as inducing expression of inflammatory cytokines, antimicrobial peptides, and growth factors. Increased expression of IL-36 cytokines has been observed in the lesional skin of patients with psoriasis. Studies of other inflammatory skin diseases have also noted similar findings, albeit to a lesser extent. IL-36 inhibition has been shown to be effective in GPP and is currently being studied for other inflammatory skin diseases. CONCLUSIONS The IL-36 pathway contributes to pathogenesis of many inflammatory skin diseases and is a promising therapeutic target.
Collapse
Affiliation(s)
- Jonwei Hwang
- University of Illinois College of Medicine, Chicago, IL, USA
| | - Jonathan Rick
- Department of Dermatology, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Jennifer Hsiao
- Department of Dermatology, University of Southern California, Los Angeles, CA, USA
| | - Vivian Y Shi
- Department of Dermatology, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| |
Collapse
|
40
|
Skin immunity: dissecting the complex biology of our body's outer barrier. Mucosal Immunol 2022; 15:551-561. [PMID: 35361906 DOI: 10.1038/s41385-022-00505-y] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 03/10/2022] [Accepted: 03/11/2022] [Indexed: 02/04/2023]
Abstract
Our skin contributes critically to health via its role as a barrier tissue, carefully regulating passage of key substrates while also providing defense against exogenous threats. Immunological processes are integral to almost every skin function and paramount to our ability to live symbiotically with skin commensal microbes and other environmental stimuli. While many parallels can be drawn to immunobiology at other mucosal sites, skin immunity demonstrates unique features that relate to its distinct topography, chemical composition and microbial ecology. Here we provide an overview of skin as an immune organ, with reference to the broader context of mucosal immunology. We review paradigms of innate as well as adaptive immune function and highlight how skin-specific structures such as hair follicles and sebaceous glands interact and contribute to these processes. Finally, we highlight for the mucosal immunology community a few emerging areas of interest for the skin immunity field moving forward.
Collapse
|
41
|
Barbieux C, Bonnet des Claustres M, Fahrner M, Petrova E, Tsoi LC, Gouin O, Leturcq F, Nicaise-Roland P, Bole C, Béziat V, Bourrat E, Schilling O, Gudjonsson JE, Hovnanian A. Netherton syndrome subtypes share IL-17/IL-36 signature with distinct IFN-α and allergic responses. J Allergy Clin Immunol 2022; 149:1358-1372. [PMID: 34543653 DOI: 10.1016/j.jaci.2021.08.024] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 08/12/2021] [Accepted: 08/27/2021] [Indexed: 12/13/2022]
Abstract
BACKGROUND Netherton syndrome (NS) is a rare recessive skin disorder caused by loss-of-function mutations in SPINK5 encoding the protease inhibitor LEKTI (lymphoepithelial Kazal-type-related inhibitor). NS patients experience severe skin barrier defects, display inflammatory skin lesions, and have superficial scaling with atopic manifestations. They present with typical ichthyosis linearis circumflexa (NS-ILC) or scaly erythroderma (NS-SE). OBJECTIVE We used a combination of several molecular profiling methods to comprehensively characterize the skin, immune cells, and allergic phenotypes of NS-ILC and NS-SE patients. METHODS We studied a cohort of 13 patients comprising 9 NS-ILC and 4 NS-SE. RESULTS Integrated multiomics revealed abnormal epidermal proliferation and differentiation and IL-17/IL-36 signatures in lesion skin and in blood in both NS endotypes. Although the molecular profiles of NS-ILC and NS-SE lesion skin were very similar, nonlesion skin of each disease subtype displayed distinctive molecular features. Nonlesion and lesion NS-SE epidermis showed activation of the type I IFN signaling pathway, while lesion NS-ILC skin differed from nonlesion NS-ILC skin by increased complement activation and neutrophil infiltration. Serum cytokine profiling and immunophenotyping of circulating lymphocytes showed a TH2-driven allergic response in NS-ILC, whereas NS-SE patients displayed mainly a TH9 axis with increased CCL22/MDC and CCL17/TARC serum levels. CONCLUSIONS This study confirms IL-17/IL-36 as the predominant signaling axes in both NS endotypes and unveils molecular features distinguishing NS-ILC and NS-SE. These results identify new therapeutic targets and could pave the way for precision medicine of NS.
Collapse
Affiliation(s)
- Claire Barbieux
- INSERM UMR 1163, Laboratory of Genetic Skin Diseases, Imagine Institute, Paris, France
| | | | - Matthias Fahrner
- Institute for Surgical Pathology, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany; Spemann Graduate School of Biology and Medicine (SGBM), Albert-Ludwigs-University Freiburg, Freiburg, Germany; Faculty of Biology, Albert-Ludwigs-University Freiburg, Freiburg, Germany
| | - Evgeniya Petrova
- INSERM UMR 1163, Laboratory of Genetic Skin Diseases, Imagine Institute, Paris, France
| | - Lam C Tsoi
- INSERM UMR 1163, Laboratory of Genetic Skin Diseases, Imagine Institute, Paris, France
| | - Olivier Gouin
- INSERM UMR 1163, Laboratory of Genetic Skin Diseases, Imagine Institute, Paris, France
| | - Florent Leturcq
- INSERM UMR 1163, Laboratory of Genetic Skin Diseases, Imagine Institute, Paris, France
| | - Pascale Nicaise-Roland
- Department of Immunology, Bichat Hospital, AP-HP-Nord, Paris, France; INSERM UMR 1152, Paris, France
| | | | - Vivien Béziat
- INSERM UMR 1163, Laboratory of Human Genetics of Infectious Diseases, Imagine Institute, Paris, France
| | | | - Oliver Schilling
- Institute for Surgical Pathology, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | | | - Alain Hovnanian
- University of Paris, Paris, France; Department of Genetics, Necker Hospital for Sick Children (AP-HP), Paris, France; Department of Dermatology, University of Michigan, Ann Arbor, Mich.
| |
Collapse
|
42
|
Chung EJ, Luo CH, Thio CLP, Chang YJ. Immunomodulatory Role of Staphylococcus aureus in Atopic Dermatitis. Pathogens 2022; 11:pathogens11040422. [PMID: 35456097 PMCID: PMC9025081 DOI: 10.3390/pathogens11040422] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 03/23/2022] [Accepted: 03/29/2022] [Indexed: 12/24/2022] Open
Abstract
Staphylococcus aureus is a gram-positive bacterium commonly found on humans, and it constitutes the skin microbiota. Presence of S. aureus in healthy individuals usually does not pose any threat, as the human body is equipped with many mechanisms to prevent pathogen invasion and infection. However, colonization of S. aureus has been correlated with many healthcare-associated infections, and has been found in people with atopic diseases. In atopic dermatitis, constant fluctuations due to inflammation of the epidermal and mucosal barriers can cause structural changes and allow foreign antigens and pathogens to bypass the first line of defense of the innate system. As they persist, S. aureus can secrete various virulence factors to enhance their survival by host invasion and evasion mechanisms. In response, epithelial cells can release damage-associated molecular patterns, or alarmins such as TSLP, IL-25, IL-33, and chemokines, to recruit innate and adaptive immune cells to cause inflammation. Until recently, IL-36 had been found to play an important role in modulating atopic dermatitis. Secretion of IL-36 from keratinocytes can activate a Th2 independent pathway to trigger symptoms of allergic reaction resulting in clinical manifestations. This mini review aims to summarize the immunomodulatory roles of S. aureus virulence factors and how they contribute to the pathogenesis of atopic diseases.
Collapse
Affiliation(s)
- Ethan Jachen Chung
- Institute of Biomedical Sciences, Academia Sinica, Taipei 115024, Taiwan; (E.J.C.); (C.-H.L.); (C.L.-P.T.)
- Graduate Institute of Microbiology, College of Medicine, National Taiwan University, Taipei 10051, Taiwan
| | - Chia-Hui Luo
- Institute of Biomedical Sciences, Academia Sinica, Taipei 115024, Taiwan; (E.J.C.); (C.-H.L.); (C.L.-P.T.)
- Graduate Institute of Microbiology, College of Medicine, National Taiwan University, Taipei 10051, Taiwan
| | - Christina Li-Ping Thio
- Institute of Biomedical Sciences, Academia Sinica, Taipei 115024, Taiwan; (E.J.C.); (C.-H.L.); (C.L.-P.T.)
| | - Ya-Jen Chang
- Institute of Biomedical Sciences, Academia Sinica, Taipei 115024, Taiwan; (E.J.C.); (C.-H.L.); (C.L.-P.T.)
- Institute of Translational Medicine and New Drug Development, China Medical University, Taichung 406040, Taiwan
- Correspondence:
| |
Collapse
|
43
|
Maiello N, Comberiati P, Giannetti A, Ricci G, Carello R, Galli E. New Directions in Understanding Atopic March Starting from Atopic Dermatitis. CHILDREN (BASEL, SWITZERLAND) 2022; 9:children9040450. [PMID: 35455494 PMCID: PMC9029734 DOI: 10.3390/children9040450] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 03/20/2022] [Accepted: 03/21/2022] [Indexed: 01/12/2023]
Abstract
Recent evidence showed that the postulated linear progression of the atopic march, from atopic dermatitis to food and respiratory allergies, does not capture the heterogeneity of allergic phenotypes, which are influenced by complex interactions between environmental, genetic, and psychosocial factors. Indeed, multiple atopic trajectories are possible in addition to the classic atopic march. Nevertheless, atopic dermatitis is often the first manifestation of an atopic march. Improved understanding of atopic dermatitis pathogenesis is warranted as this could represent a turning point in the prevention of atopic march. In this review, we outline the recent findings on the pathogenetic mechanisms leading to atopic dermatitis that could be targeted by intervention strategies for the prevention of atopic march.
Collapse
Affiliation(s)
- Nunzia Maiello
- Department of Woman, Child and General and Specialized Surgery, University of Campania “Luigi Vanvitelli”, 81100 Naples, Italy
- Correspondence:
| | - Pasquale Comberiati
- Department of Clinical and Experimental Medicine, Section of Pediatrics, University of Pisa, 56126 Pisa, Italy;
- Department of Clinical Immunology and Allergology, I.M. Sechenov First Moscow State Medical University, 119991 Moscow, Russia
| | - Arianna Giannetti
- Pediatric Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy;
| | - Giampaolo Ricci
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40138 Bologna, Italy;
| | - Rossella Carello
- Pediatric Allergic Unit, S.Pietro Hospital FbF Roma, 00189 Rome, Italy; (R.C.); (E.G.)
| | - Elena Galli
- Pediatric Allergic Unit, S.Pietro Hospital FbF Roma, 00189 Rome, Italy; (R.C.); (E.G.)
| |
Collapse
|
44
|
Frühbeck G, Gómez-Ambrosi J, Ramírez B, Mentxaka A, Rodríguez A, Becerril S, Reina G, Valentí V, Moncada R, Silva C, Catalán V. Increased Levels of Interleukin-36 in Obesity and Type 2 Diabetes Fuel Adipose Tissue Inflammation by Inducing Its Own Expression and Release by Adipocytes and Macrophages. Front Immunol 2022; 13:832185. [PMID: 35222417 PMCID: PMC8863603 DOI: 10.3389/fimmu.2022.832185] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Accepted: 01/20/2022] [Indexed: 02/03/2023] Open
Abstract
Interleukin (IL)-36 is a recently described cytokine with well-known functions in the regulation of multiple inflammatory diseases. Since no data exists on how this cytokine regulates adipose tissue (AT) homeostasis, we aimed to explore the function of a specific isoform, IL-36γ, an agonist, in human obesity and obesity-associated type 2 diabetes as well as in AT inflammation and fibrosis. Plasma IL-36γ was measured in 91 participants in a case-control study and the effect of weight loss was evaluated in 31 patients with severe obesity undergoing bariatric surgery. Gene expression levels of IL36G and its receptor were analyzed in relevant human metabolic tissues. The effect of inflammatory factors and IL-36γ was determined in vitro in human adipocytes and macrophages. We found, for the first time, that the increased (P<0.05) circulating levels of IL-36γ in patients with obesity decreased (P<0.001) after weight and fat loss achieved by Roux-en-Y gastric bypass and that gene expression levels of IL36G were upregulated in the visceral AT (P<0.05) and in the peripheral blood mononuclear cells (P<0.01) from patients with obesity. We also demonstrated increased (P<0.05) expression levels of Il36g in the epididymal AT from diet-induced obese mice. IL36G was significantly enhanced (P<0.001) by LPS in human adipocytes and monocyte-derived macrophages, while no changes were found after the incubation with anti-inflammatory cytokines. The addition of IL-36γ for 24 h strongly induced (P<0.01) its own expression as well as key inflammatory and chemoattractant factors with no changes in genes associated with fibrosis. Furthermore, adipocyte-conditioned media obtained from patients with obesity increased (P<0.01) the release of IL-36γ and the expression (P<0.05) of cathepsin G (CTSG) in monocyte-derived macrophages. These findings provide, for the first time, evidence about the properties of IL-36γ in the regulation of AT-chronic inflammation, emerging as a link between AT biology and the obesity-associated comorbidities.
Collapse
Affiliation(s)
- Gema Frühbeck
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Pamplona, Spain,CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Pamplona, Spain,Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain,Department of Endocrinology and Nutrition, Clínica Universidad de Navarra, Pamplona, Spain,*Correspondence: Victoria Catalán, ; Gema Frühbeck,
| | - Javier Gómez-Ambrosi
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Pamplona, Spain,CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Pamplona, Spain,Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Beatriz Ramírez
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Pamplona, Spain,CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Pamplona, Spain,Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Amaia Mentxaka
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Pamplona, Spain,CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Pamplona, Spain
| | - Amaia Rodríguez
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Pamplona, Spain,CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Pamplona, Spain,Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Sara Becerril
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Pamplona, Spain,CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Pamplona, Spain,Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain
| | - Gabriel Reina
- Department of Microbiology, Clínica Universidad de Navarra, Pamplona, Spain
| | - Victor Valentí
- Department of Surgery, Clínica Universidad de Navarra, Pamplona, Spain
| | - Rafael Moncada
- Department of Anesthesia, Clínica Universidad de Navarra, Pamplona, Spain
| | - Camilo Silva
- Department of Endocrinology and Nutrition, Clínica Universidad de Navarra, Pamplona, Spain
| | - Victoria Catalán
- Metabolic Research Laboratory, Clínica Universidad de Navarra, Pamplona, Spain,CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Pamplona, Spain,Obesity and Adipobiology Group, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, Spain,*Correspondence: Victoria Catalán, ; Gema Frühbeck,
| |
Collapse
|
45
|
Luo X, Chen J, Yang H, Hu X, Alphonse MP, Shen Y, Kawakami Y, Zhou X, Tu W, Kawakami T, Wan M, Archer NK, Wang H, Gao P. Dendritic cell immunoreceptor drives atopic dermatitis by modulating oxidized CaMKII-involved mast cell activation. JCI Insight 2022; 7:152559. [PMID: 35113811 PMCID: PMC8983143 DOI: 10.1172/jci.insight.152559] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Accepted: 01/28/2022] [Indexed: 11/17/2022] Open
Abstract
Allergens have been identified as potential triggers in patients with atopic dermatitis (AD). AD patients are highly sensitive to cockroach allergen. The underlying mechanism, however, remains undetermined. Here, we established a cockroach allergen-induced AD-like mouse model and demonstrated that repeated exposure to cockroach allergen led to aggravated mouse skin inflammation, characterized by increased type 2 immunity, type 2 innate lymphoid cells (ILC2s), and mast cells. Increased skin mast cells were also observed in AD patients. AD mice with mast cell-deficient mice (kitW-sh/W-sh) showed diminished skin inflammation, suggesting that mast cells are required in allergen-induced skin inflammation. Furthermore, dendritic cell immuno-receptor (DCIR) is up-regulated in skin mast cells of AD patients and mediates allergen binding and uptake. DCIR-/- mice or reconstituted kitW-sh/W-sh mice with DCIR-/- mast cells showed a significant reduction in AD-like inflammation. Both in vitro and in vivo analyses demonstrated that DCIR-/- mast cells had reduced IgE-mediated mast cell activation and passive cutaneous anaphylaxis. Mechanistically, DCIR regulates allergen-induced IgE-mediated mast cell ROS generation and oxidation of calmodulin kinase II (ox-CaMKII). ROS-resistant CaMKII (MM-VVδ) prevents allergen-induced mast cell activation and inflammatory mediator release. Our study reveals a previously unrecognized DCIR-ROS-CaMKII axis that controls allergen-induced mast cell activation and AD-like inflammation.
Collapse
Affiliation(s)
- Xiaoyan Luo
- Pediatric Dermatology, Chongqing Medical University, Chongqing, China
| | - Jingsi Chen
- Pediatric Dermatology, Chongqing Medical University, Chongqing, China
| | - Huan Yang
- Pediatric Dermatology, Chongqing Medical University, Chongqing, China
| | - Xinyue Hu
- Division of Allergy and Clinical Immunology, Johns Hopkins University School of Medicine, Baltimore, United States of America
| | - Martin P Alphonse
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, United States of America
| | - Yingchun Shen
- Division of Allergy and Clinical Immunology, Johns Hopkins University School of Medicine, Baltimore, United States of America
| | - Yuko Kawakami
- Division of Cell Biology, La Jolla Institute for Allergy and Immunology, La Jolla, United States of America
| | - Xiaoying Zhou
- Pediatric Dermatology, Chongqing Medical University, Chongqing, China
| | - Wei Tu
- Division of Allergy and Clinical Immunology, Johns Hopkins University School of Medicine, Baltimore, United States of America
| | - Toshiaki Kawakami
- Division of Cell Biology, La Jolla Institute for Allergy and Immunology, La Jolla, United States of America
| | - Mei Wan
- Johns Hopkins University School of Medicine, Baltimore, United States of America
| | - Nathan K Archer
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, United States of America
| | - Hua Wang
- Pediatric Dermatology, Chongqing Medical University, Chongqing, China
| | - Peisong Gao
- Division of Allergy and Clinical Immunology, Johns Hopkins University School of Medicine, Baltimore, United States of America
| |
Collapse
|
46
|
Keet CA, Berin MC. The year in food allergy. J Allergy Clin Immunol 2022; 149:867-873. [PMID: 35031274 DOI: 10.1016/j.jaci.2021.12.785] [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: 11/11/2021] [Revised: 12/28/2021] [Accepted: 12/31/2021] [Indexed: 11/16/2022]
Abstract
Research into food allergy continues to rapidly evolve, accompanying and driving real changes in the clinical approach to these diseases. The past year has seen the rollout of the first treatment approved for active management of food allergy, more data on alternative methods of treatment, the continued evolution of strategies for prevention of food allergy, a renewed interest in phenotyping food allergy subtypes, and, importantly, key new insights into the pathophysiology of food allergy. We expect that in the coming years, the therapies that are in preclinical or early clinical evaluation now will make their way to the clinic, finally allowing the possibility of safe and effective treatments for food allergy.
Collapse
Affiliation(s)
- Corinne A Keet
- Department of Pediatrics, School of Medicine, University of North Carolina, Chapel Hill, NC.
| | - M Cecilia Berin
- Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY
| |
Collapse
|
47
|
Macleod T, Berekmeri A, Bridgewood C, Stacey M, McGonagle D, Wittmann M. The Immunological Impact of IL-1 Family Cytokines on the Epidermal Barrier. Front Immunol 2022; 12:808012. [PMID: 35003136 PMCID: PMC8733307 DOI: 10.3389/fimmu.2021.808012] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 12/06/2021] [Indexed: 12/25/2022] Open
Abstract
The skin barrier would not function without IL-1 family members, but their physiological role in the immunological aspects of skin barrier function are often overlooked. This review summarises the role of IL-1 family cytokines (IL-1α, IL-1β, IL-1Ra, IL-18, IL-33, IL-36α, IL-36β, IL-36γ, IL-36Ra, IL-37 and IL-38) in the skin. We focus on novel aspects of their interaction with commensals and pathogens, the important impact of proteases on cytokine activity, on healing responses and inflammation limiting mechanisms. We discuss IL-1 family cytokines in the context of IL-4/IL-13 and IL-23/IL-17 axis-driven diseases and highlight consequences of human loss/gain of function mutations in activating or inhibitory pathway molecules. This review highlights recent findings that emphasize the importance of IL-1 family cytokines in both physiological and pathological cutaneous inflammation and emergent translational therapeutics that are helping further elucidate these cytokines.
Collapse
Affiliation(s)
- Tom Macleod
- School of Molecular and Cellular Biology, University of Leeds, Leeds, United Kingdom.,Leeds Institute of Rheumatic and Musculoskeletal Medicine (LIRMM), University of Leeds, Leeds, United Kingdom
| | - Anna Berekmeri
- Leeds Institute of Rheumatic and Musculoskeletal Medicine (LIRMM), University of Leeds, Leeds, United Kingdom
| | - Charlie Bridgewood
- Leeds Institute of Rheumatic and Musculoskeletal Medicine (LIRMM), University of Leeds, Leeds, United Kingdom
| | - Martin Stacey
- School of Molecular and Cellular Biology, University of Leeds, Leeds, United Kingdom
| | - Dennis McGonagle
- Leeds Institute of Rheumatic and Musculoskeletal Medicine (LIRMM), University of Leeds, Leeds, United Kingdom.,National Institute for Health Research (NIHR) Leeds Biomedical Research Centre (BRC), The Leeds Teaching Hospitals, Leeds, United Kingdom
| | - Miriam Wittmann
- Leeds Institute of Rheumatic and Musculoskeletal Medicine (LIRMM), University of Leeds, Leeds, United Kingdom.,National Institute for Health Research (NIHR) Leeds Biomedical Research Centre (BRC), The Leeds Teaching Hospitals, Leeds, United Kingdom
| |
Collapse
|
48
|
Abstract
Microbes in the 21st century are understood as symbionts ‘completing’ the human ‘superorganism’ (Homo sapiens plus microbial partners-in-health). This paper addresses a significant paradox: despite the vast majority of our genes being microbial, the lack of routine safety testing for the microbiome has led to unintended collateral side effects from pharmaceuticals that can damage the microbiome and inhibit innate ‘colonization resistance’ against pathobionts. Examples are discussed in which a Microbiome First Medicine approach provides opportunities to ‘manage our microbes’ holistically, repair dysbiotic superorganisms, and restore health and resilience in the gut and throughout the body: namely, managing nosocomial infections for Clostridioides difficile and Staphylococcus aureus and managing the gut and neural systems (gut–brain axis) in autism spectrum disorder. We then introduce a risk analysis tool: the evidence map. This ‘mapping’ tool was recently applied by us to evaluate evidence for benefits, risks, and uncertainties pertaining to the breastmilk ecosystem. Here, we discuss the potential role of the evidence map as a risk analysis methodology to guide scientific and societal efforts to: (1) enhance ecosystem resilience, (2) ‘manage our microbes’, and (3) minimize the adverse effects of both acute and chronic diseases.
Collapse
|