1
|
Kapitány A, Soltész L, Stercel V, Szabó L, Somogyi O, Janka EA, Nagy V, Póliska S, Gáspár K, Hendrik Z, Törőcsik D, Dajnoki Z, Szegedi A. Chronological maturation of the skin immune barrier is topographically different. Mucosal Immunol 2025; 18:730-741. [PMID: 40158778 DOI: 10.1016/j.mucimm.2025.03.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 03/06/2025] [Accepted: 03/25/2025] [Indexed: 04/02/2025]
Abstract
Adult skin varies across regions, with differences in chemical, physical, microbiota, and immune barriers. However, data on topographical immune differences in other age groups are limited. This study aimed to explore the chronological maturation of the immune barrier in various skin regions. A TaqMan low-density array and immunohistochemical and immunofluorescence detection of various immune cells and mediators in sebaceous gland-rich (SGR) and gland-poor (GP) healthy skin were performed in children, adolescents, and adults. The maturation of SGR skin showed a general upward trend in the mRNA levels of most Th17-related molecules with a significant increase in IL-1B from childhood to adulthood, but with only a slight elevation between childhood and adolescence. In addition, T cell, Treg, dendritic cell (DC) counts, as well as the levels of several Th17-related proteins (IL-17, IL-10, IL-23, CCL20, S100A8, sfTSLP, LCN2), increased significantly with age. In GP skin, AHR mRNA levels decreased, while Th17-related protein levels increased, although only moderately. When comparing the two regions, SGR and GP skin were similar in childhood, with differences emerging in adolescence and becoming significant in adulthood, particularly in the IL-17 pathway, mainly produced by Th17 cells. Our results show a similarly directed maturation process in GP and SGR regions, with more pronounced development of the SGR skin immune barrier (with more immune cell infiltration and cytokine production) during and after the adolescence. This is likely to be related to the significant changes in the chemical and microbiota barriers of the SGR skin during adolescence, and may explain the high incidence of inflammatory skin diseases on the SGR skin of adolescents, highlighting the need for targeted skin care in this region.
Collapse
Affiliation(s)
- Anikó Kapitány
- Department of Dermatology, Center of Excellence, Faculty of Medicine, University of Debrecen, 98. Nagyerdei Krt. Debrecen H-4032, Hungary; HUN-REN-DE Allergology Research Group, 98. Nagyerdei Krt. Debrecen H-4032, Hungary
| | - Lilla Soltész
- Department of Dermatology, Center of Excellence, Faculty of Medicine, University of Debrecen, 98. Nagyerdei Krt. Debrecen H-4032, Hungary; Gyula Petrányi Doctoral School of Allergy and Clinical Immunology, University of Debrecen, 98. Nagyerdei Krt. Debrecen H-4032, Hungary
| | - Vivien Stercel
- Department of Pediatrics, Faculty of Medicine, University of Debrecen, 98. Nagyerdei Krt. Debrecen H-4032, Hungary
| | - Lilla Szabó
- Department of Dermatology, Center of Excellence, Faculty of Medicine, University of Debrecen, 98. Nagyerdei Krt. Debrecen H-4032, Hungary; Gyula Petrányi Doctoral School of Allergy and Clinical Immunology, University of Debrecen, 98. Nagyerdei Krt. Debrecen H-4032, Hungary
| | - Orsolya Somogyi
- Department of Dermatology, Center of Excellence, Faculty of Medicine, University of Debrecen, 98. Nagyerdei Krt. Debrecen H-4032, Hungary; Gyula Petrányi Doctoral School of Allergy and Clinical Immunology, University of Debrecen, 98. Nagyerdei Krt. Debrecen H-4032, Hungary
| | - Eszter Anna Janka
- Department of Dermatology, Center of Excellence, Faculty of Medicine, University of Debrecen, 98. Nagyerdei Krt. Debrecen H-4032, Hungary; HUN-REN-DE Allergology Research Group, 98. Nagyerdei Krt. Debrecen H-4032, Hungary
| | - Viktória Nagy
- Department of Dermatology, Center of Excellence, Faculty of Medicine, University of Debrecen, 98. Nagyerdei Krt. Debrecen H-4032, Hungary; Gyula Petrányi Doctoral School of Allergy and Clinical Immunology, University of Debrecen, 98. Nagyerdei Krt. Debrecen H-4032, Hungary
| | - Szilárd Póliska
- Genomic Medicine and Bioinformatics Core Facility, Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, 98. Nagyerdei Krt. Debrecen H-4032, Hungary
| | - Krisztián Gáspár
- Department of Dermatology, Center of Excellence, Faculty of Medicine, University of Debrecen, 98. Nagyerdei Krt. Debrecen H-4032, Hungary; HUN-REN-DE Allergology Research Group, 98. Nagyerdei Krt. Debrecen H-4032, Hungary
| | - Zoltán Hendrik
- Department of Forensic Medicine, Faculty of Medicine, University of Debrecen, 98. Nagyerdei Krt. Debrecen H-4032, Hungary
| | - Dániel Törőcsik
- Department of Dermatology, Center of Excellence, Faculty of Medicine, University of Debrecen, 98. Nagyerdei Krt. Debrecen H-4032, Hungary; HUN-REN-DE Allergology Research Group, 98. Nagyerdei Krt. Debrecen H-4032, Hungary
| | - Zsolt Dajnoki
- Department of Dermatology, Center of Excellence, Faculty of Medicine, University of Debrecen, 98. Nagyerdei Krt. Debrecen H-4032, Hungary; HUN-REN-DE Allergology Research Group, 98. Nagyerdei Krt. Debrecen H-4032, Hungary
| | - Andrea Szegedi
- Department of Dermatology, Center of Excellence, Faculty of Medicine, University of Debrecen, 98. Nagyerdei Krt. Debrecen H-4032, Hungary; HUN-REN-DE Allergology Research Group, 98. Nagyerdei Krt. Debrecen H-4032, Hungary.
| |
Collapse
|
2
|
Kalankariyan S, Thottapillil A, Saxena A, Srivatsn S M, Kadamkode V, Kapoor R, Mitra R, Raut J, Venkatesh KV. An in silico approach deciphering the commensal dynamics in the cutaneous milieu. NPJ Syst Biol Appl 2025; 11:42. [PMID: 40335508 PMCID: PMC12058978 DOI: 10.1038/s41540-025-00524-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Accepted: 04/14/2025] [Indexed: 05/09/2025] Open
Abstract
The skin microbiota, particularly coagulase-negative staphylococci (CoNS) such as S. epidermidis, plays a crucial role in maintaining skin health and immunity. S. epidermidis, a predominant commensal species, interacts intimately with keratinocytes to regulate immune responses and antimicrobial defence mechanisms. Metabolic byproducts like short-chain fatty acids (SCFAs) influence keratinocyte activation, while cell wall components engage Toll-like receptors (TLRs) to modulate inflammation. These interactions are fundamental for preserving skin homeostasis and combating pathogenic invaders. Our comprehensive mathematical model, integrating commensal dynamics, immune responses, and skin microenvironment variables, provides insights into these intricate interactions. The model delves into the complexities of skin scenarios and perturbations, aiming to understand the colonization dynamics of S. epidermidis and its influence on skin barrier functions. It examines how disruptions in key factors such as AMP, growth factor-mediated repair pathways, and filaggrin mutations influence the behaviour of the system. The study depicts the skin microenvironment as a highly dynamic one, highlighting the critical role of S. epidermidis and capturing its role in barrier dysfunction caused by internal and external factors. By offering insights into skin barrier function and immune responses, the model illuminates key interactions of commensals within the skin microenvironment which can ultimately benefit skin health.
Collapse
Affiliation(s)
| | | | - Abha Saxena
- MetFlux Research Private Limited, Bengaluru, India
| | | | | | | | | | | | - K V Venkatesh
- MetFlux Research Private Limited, Bengaluru, India.
- Department of Chemical Engineering, Indian Institute of Technology Bombay, Mumbai, Maharashtra, India.
| |
Collapse
|
3
|
Thorstenson JC, Horswill AR. Staphylococcus aureus SspA (V8 protease): New skin pathogenesis insights into an old enzyme. PLoS Pathog 2025; 21:e1013048. [PMID: 40273042 PMCID: PMC12021127 DOI: 10.1371/journal.ppat.1013048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/26/2025] Open
Affiliation(s)
- John C. Thorstenson
- Department of Immunology & Microbiology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States of America
| | - Alexander R. Horswill
- Department of Immunology & Microbiology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States of America
- Department of Veterans Affairs, Eastern Colorado Health Care System, Aurora, Colorado, United States of America
| |
Collapse
|
4
|
Gallais Sérézal I, Kirma J, Sarkar MK, Cole C, Xing X, Bogle R, Fox J, Coon A, vanStraalen KR, Dobry C, Xu LH, Kahlenberg JM, Harms PW, Billi AC, Tsoi LC, Giacani L, Gudjonsson JE. Characterizing the immune infiltrate in secondary syphilis: implications for transmission and pathology. Front Immunol 2025; 16:1549206. [PMID: 40201184 PMCID: PMC11975926 DOI: 10.3389/fimmu.2025.1549206] [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: 12/20/2024] [Accepted: 02/14/2025] [Indexed: 04/10/2025] Open
Abstract
Introduction Syphilis is a complex disease with variable clinical presentation where symptomatic and potentially infectious stages alternate with periods of latency, representing a fascinating model to study immune evasion and host immune responses. Methods Immunohistochemistry (IHC), bulk, and single-cell RNA sequencing were performed on formalin-fixed paraffin-embedded skin biopsies collected from subjects with secondary syphilis. Additionally, PBMCs from healthy individuals and either primary or MyD88 knock-out keratinocytes were exposed to live Treponema pallidum cells to define initial skin responses to the bacteria. Results Immunohistochemistry of secondary syphilis skin lesions showed a polymorphous immune infiltrate with colocalization of T cells, B cells and antigen-presenting cells. Single-cell analysis revealed distinct cellular contributions to the immune response, with prominent immune-stromal crosstalk accompanied by altered keratinocyte differentiation and decreased intraepidermal communication. Notably, prominent inflammatory signals were countered by concomitant regulatory responses, particularly in infiltrating myeloid cells. Exposure of PBMCs to live T. pallidum inhibited immune responses, while exposure to sonicated cells triggered CXCL1 and CXCL3 upregulation. Keratinocytes responded to both intact and sonicated T. pallidum with upregulation of type-I interferon responses that, however, were abolished in MYD88-deficient but not in STING-deficient keratinocytes. Discussion Our data provide novel insights into the contribution of epidermal TLR sensing through MYD88 to the host response to syphilis infection, highlighting mechanisms by which T. pallidum evades immune responses in skin that may facilitate transmission of this pathogen through the skin.
Collapse
Affiliation(s)
- Irène Gallais Sérézal
- Department of Dermatology, Institut National de la Santé et de la Recherche Médicale (INSERM) 1098, Franche Comté University, Besançon University Hospital, Besançon, France
- Department of Dermatology, University of Michigan, Ann Arbor, MI, United States
| | - Joseph Kirma
- Department of Dermatology, University of Michigan, Ann Arbor, MI, United States
| | - Mrinal K. Sarkar
- Department of Dermatology, University of Michigan, Ann Arbor, MI, United States
| | - Christopher Cole
- Department of Dermatology, University of Michigan, Ann Arbor, MI, United States
| | - Xianying Xing
- Department of Dermatology, University of Michigan, Ann Arbor, MI, United States
| | - Rachael Bogle
- Laboratory for Experimental Immunodermatology, Department of Dermatology, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Jennifer Fox
- Department of Dermatology, University of Michigan, Ann Arbor, MI, United States
| | - Anthony Coon
- Department of Dermatology, University of Michigan, Ann Arbor, MI, United States
| | - Kelsey R. vanStraalen
- Laboratory for Experimental Immunodermatology, Department of Dermatology, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Craig Dobry
- Department of Dermatology, University of Michigan, Ann Arbor, MI, United States
| | - Linda H. Xu
- Department of Medicine, Division of Allergy and Infectious Diseases, University of Washington, Seattle, WA, United States
| | - J. Michelle Kahlenberg
- Department of Internal Medicine, Division of Rheumatology, University of Michigan, Ann Arbor, MI, United States
| | - Paul W. Harms
- Department of Pathology, University of Michigan, Ann Arbor, MI, United States
| | - Allison C. Billi
- Department of Dermatology, University of Michigan, Ann Arbor, MI, United States
| | - Lam C. Tsoi
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, United States
| | - Lorenzo Giacani
- Department of Medicine, Division of Allergy and Infectious Diseases, University of Washington, Seattle, WA, United States
| | - Johann E. Gudjonsson
- Department of Dermatology, University of Michigan, Ann Arbor, MI, United States
- Department of Internal Medicine, Division of Rheumatology, University of Michigan, Ann Arbor, MI, United States
| |
Collapse
|
5
|
Abida O, Ramiro R, Bahloul E, Frikha R, Charfi S, Turki H, Gonçalves CP, Masmoudi H. Distinct anti-microbial peptides expression patterns and microbiome profiles in skin of Tunisian endemic Pemphigus foliaceus patients. Arch Dermatol Res 2025; 317:497. [PMID: 40009223 DOI: 10.1007/s00403-025-04000-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2024] [Revised: 01/31/2025] [Accepted: 02/03/2025] [Indexed: 02/27/2025]
Abstract
Pemphigus foliaceus (PF) is a multifactorial skin disease. Substantial evidence for microbiota dysbiosis in skin disorders was gradually revealed. In PF patients' skin lesions, we characterized the profile of microbial communities and the expression of microbial peptides. Using real-time reverse transcriptase PCR and immunohistochemistry, skin lesions were analyzed for gene and protein expression of human β-defensin (hBD) 1, 2, and 3, cathelicidin (LL-37), RNAse-7, and psoriasin. Bacterial 16S rRNA gene sequencing was used for assessing skin microbial communities in 15 samples from PF patients' lesioned skin and 11 PF patients' non-lesioned skin. Gene expression of hBD 2 and 3 and psoriasin were significantly downregulated in skin samples from remittent patients compared to chronic or de novo diagnosed patients. Protein expression of hBD 2, Psoriasin, and LL-37 was increased in skin from de novo patients compared to skin from healthy donors showing markedly different distribution patterns. The skin microbial analysis revealed a substantial difference in microbiome diversity between lesioned and non-lesioned skin of de novo PF patients and, non-lesioned skin of remittent patients. In addition, microbiome diversity within samples of lesioned skin from de novo PF patients showed lower diversity with a lower abundance of specific bacterial genera, namely Dermabacter, Psychrobacter, and Bradyrhizobium. Thus, there is a noticeable over-representation of Staphylococcus and decreased richness in the bacterial communities of PF-active skin lesions. Our data supports the hypothesis that active skin lesions in PF patients exhibit alterations in skin bacterial diversity interlinked with increased expression of AMPs.
Collapse
Affiliation(s)
- Olfa Abida
- "Autoimmunity, Cancer and Immunogenetics" research laboratory (LR18SP12), Immunology Department, Habib Bourguiba Hospital, University of Sfax, Sfax, 3029, Tunisia.
| | | | - Emna Bahloul
- Dermatology Department, Hedi Chaker Hospital, University of Sfax, Sfax, Tunisia
| | - Rim Frikha
- "Autoimmunity, Cancer and Immunogenetics" research laboratory (LR18SP12), Immunology Department, Habib Bourguiba Hospital, University of Sfax, Sfax, 3029, Tunisia
| | - Slim Charfi
- Anatomy and Pathology Department, Habib Bourguiba Hospital, Sfax, Tunisia
| | - Hamida Turki
- Dermatology Department, Hedi Chaker Hospital, University of Sfax, Sfax, Tunisia
| | | | - Hatem Masmoudi
- "Autoimmunity, Cancer and Immunogenetics" research laboratory (LR18SP12), Immunology Department, Habib Bourguiba Hospital, University of Sfax, Sfax, 3029, Tunisia
| |
Collapse
|
6
|
Dajnoki Z, Kapitány A, Eyerich K, Eyerich S, Törőcsik D, Szegedi A. Topographical variations in the skin barrier and their role in disease pathogenesis. J Eur Acad Dermatol Venereol 2024. [PMID: 39607016 DOI: 10.1111/jdv.20463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Accepted: 10/09/2024] [Indexed: 11/29/2024]
Abstract
The skin barrier can be divided into at least four functional units: chemical, microbial, physical and immunological barriers. The chemical and microbial barriers have previously been shown to exhibit different characteristics in topographically distinct skin regions. There is increasing evidence that the physical and immunological barriers also show marked variability in different areas of the skin. Here, we review recent data on the topographical variations of skin barrier components, the contribution of these variations to the homeostatic function of the skin and their impact on the pathogenesis of specific immune-mediated skin diseases (such as atopic dermatitis and papulopustular rosacea). Recognition of these topographical barrier differences will improve our understanding of skin homeostasis and disease pathogenesis and provide a basis for body site-specific targeted therapies.
Collapse
Affiliation(s)
- Z Dajnoki
- Department of Dermatology, MTA Centre of Excellence, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
- HUN-REN-UD Allergology Research Group, Debrecen, Hungary
| | - A Kapitány
- Department of Dermatology, MTA Centre of Excellence, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
- HUN-REN-UD Allergology Research Group, Debrecen, Hungary
| | - K Eyerich
- Department of Dermatology and Venerology, Medical Center, University of Freiburg, Freiburg, Germany
| | - S Eyerich
- Center for Allergy and Environment (ZAUM), Technical University and Helmholtz Center Munich, Munich, Germany
| | - D Törőcsik
- Department of Dermatology, MTA Centre of Excellence, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
- HUN-REN-UD Allergology Research Group, Debrecen, Hungary
| | - A Szegedi
- Department of Dermatology, MTA Centre of Excellence, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
- HUN-REN-UD Allergology Research Group, Debrecen, Hungary
| |
Collapse
|
7
|
Criado PR, Miot HA, Bueno-Filho R, Ianhez M, Criado RFJ, de Castro CCS. Update on the pathogenesis of atopic dermatitis. An Bras Dermatol 2024; 99:895-915. [PMID: 39138034 PMCID: PMC11551276 DOI: 10.1016/j.abd.2024.06.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Accepted: 06/18/2024] [Indexed: 08/15/2024] Open
Abstract
Atopic dermatitis is a chronic, recurrent, and multifactorial skin-mucosal manifestation resulting from the interaction between elements mainly associated with the skin barrier deficit, the homeostasis of the immune response, neurological aspects, and patterns of reactivity to environmental antigens, which are established in genetically predisposed individuals. In addition to the skin, atopic diathesis involves other organs such as the airways (upper and lower), eyes, digestive tract, and neuropsychiatric aspects, which inflict additional morbidity on the dermatological patient. The different phenotypes of the disease fundamentally depend on the participation of each of these factors, in different life circumstances, such as age groups, occupational exposure patterns, physical activity, pollution, genetic load, and climatic factors. A better understanding of the complexity of its pathogenesis allows not only the understanding of therapeutic targets but also how to identify preponderant elements that mediate disease activity in each circumstance, for selecting the best treatment strategies and mitigation of triggering factors. This narrative review presents an update on the pathogenesis of atopic dermatitis, especially aimed at understanding the clinical manifestations, the main disease phenotypes and the context of available therapeutic strategies.
Collapse
Affiliation(s)
- Paulo Ricardo Criado
- Centro Universitário Faculdade de Medicina do ABC, Santo André, SP, Brazil; Faculdade de Ciências Médicas de Santos (Centro Universitário Lusíada), Santos, SP, Brazil.
| | - Hélio Amante Miot
- Department of Dermatology, Faculdade de Medicina de Botucatu, Universidade do Estado de São Paulo, Botucatu, SP, Brazil
| | - Roberto Bueno-Filho
- Division of Dermatology, Department of Internal Medicine, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, SP, Brazil
| | - Mayra Ianhez
- Department of Dermatology, Hospital de Doenças Tropicais de Goiás, Goiânia, GO, Brazil
| | - Roberta Fachini Jardim Criado
- Centro Universitário Faculdade de Medicina do ABC, Santo André, SP, Brazil; Alergoskin Alergia e Dermatologia, UCARE Center and ADCARE, Santo André, SP, Brazil
| | - Caio César Silva de Castro
- Pontifícia Universidade Católica do Paraná, Curitiba, PR, Brazil; Hospital de Dermatologia Sanitária do Paraná, Curitiba, PR, Brazil
| |
Collapse
|
8
|
Cervantes Recalde MF, Schmidt J, Girardi C, Massironi M, Rechl ML, Hans J, Stuhlmann D, Somoza V, Lieder B. Capsaicin attenuates the effect of inflammatory cytokines in a HaCaT cell model for basal keratinocytes. Front Pharmacol 2024; 15:1474898. [PMID: 39469627 PMCID: PMC11513304 DOI: 10.3389/fphar.2024.1474898] [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: 08/02/2024] [Accepted: 09/25/2024] [Indexed: 10/30/2024] Open
Abstract
Introduction The resolution of the skin's inflammatory response is only possible if its barrier function is restored. TRPV1 channel activation plays an important role during inflammation but the effect of this activation on the skin barrier under inflammatory conditions has not been clarified. We hypothesize that it could potentially aid the keratinocyte barrier by reducing inflammatory cytokine release and promoting tight junction development. Methods To explore the role of TRPV1 activation in inflammation, we designed and optimized an in vitro model of keratinocytes with basal epidermal layer characteristics using HaCaT cells and TNFα to induce inflammation. Results TNFα increased the gene expression of tight junction protein claudin 1 (CLDN1) by at least 2.60 ± 0.16-fold, in a concentration-dependent manner, over a 48 h period. The administration of a capsaicin pre-treatment reduced the CLDN1 expression to 1.51 ± 0.16-fold during the first 6 h after TNFα induction, whereas IL-8 cytokine release was reduced 0.64 ± 0.17-fold. After 48 h, CLDN1 protein levels increased by a factor of 6.57 ± 1.39 compared to cells only treated with TNFα. Discussion These results suggest that activation of TRPV1 by capsaicin can potentiate the increase in CLDN1 expression and CLDN1 protein synthesis induced by TNFα in cultured keratinocytes, while reducing the release of IL-8.
Collapse
Affiliation(s)
- Maria Fernanda Cervantes Recalde
- Institute of Physiological Chemistry, Faculty of Chemistry, University of Vienna, Vienna, Austria
- Vienna Doctoral School in Chemistry (DoSChem), University of Vienna, Vienna, Austria
| | - Jana Schmidt
- Institute of Physiological Chemistry, Faculty of Chemistry, University of Vienna, Vienna, Austria
| | | | | | - Markus Leo Rechl
- Institute of Physiological Chemistry, Faculty of Chemistry, University of Vienna, Vienna, Austria
- Vienna Doctoral School in Chemistry (DoSChem), University of Vienna, Vienna, Austria
- Christian Doppler Laboratory for Taste Research, Faculty of Chemistry, University of Vienna, Vienna, Austria
| | | | | | - Veronika Somoza
- Institute of Physiological Chemistry, Faculty of Chemistry, University of Vienna, Vienna, Austria
- Leibniz Institute of Food Systems Biology, Technical University of Munich, Freising, Germany
| | - Barbara Lieder
- Institute of Physiological Chemistry, Faculty of Chemistry, University of Vienna, Vienna, Austria
- Christian Doppler Laboratory for Taste Research, Faculty of Chemistry, University of Vienna, Vienna, Austria
- Institute of Clinical Nutrition, University of Hohenheim, Stuttgart, Germany
| |
Collapse
|
9
|
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] [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
|
10
|
Petronio Petronio G, Di Naro M, Venditti N, Guarnieri A, Cutuli MA, Magnifico I, Medoro A, Foderà E, Passarella D, Nicolosi D, Di Marco R. Targeting S. aureus Extracellular Vesicles: A New Putative Strategy to Counteract Their Pathogenic Potential. Pharmaceutics 2024; 16:789. [PMID: 38931910 PMCID: PMC11207539 DOI: 10.3390/pharmaceutics16060789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 05/27/2024] [Accepted: 06/07/2024] [Indexed: 06/28/2024] Open
Abstract
Long-term inflammatory skin disease atopic dermatitis is characterized by dry skin, itching, and eczematous lesions. During inflammation skin barrier protein impairment promotes S. aureus colonisation in the inflamed skin, worsening AD patient's clinical condition. Proteomic analysis revealed the presence of several immune evasion proteins and virulence factors in S. aureus extracellular vesicles (EVs), suggesting a possible role for these proteins in the pathophysiology of atopic dermatitis. The objective of this study is to assess the efficacy of a wall fragment obtained from a patented strain of C. acnes DSM28251 (c40) and its combination with a mucopolysaccharide carrier (HAc40) in counteract the pathogenic potential of EVs produced by S. aureus ATCC 14458. Results obtained from in vitro studies on HaCaT keratinocyte cells showed that HAc40 and c40 treatment significantly altered the size and pathogenicity of S. aureus EVs. Specifically, EVs grew larger, potentially reducing their ability to interact with the target cells and decreasing cytotoxicity. Additionally, the overexpression of the tight junctions mRNA zona occludens 1 (ZO1) and claudin 1 (CLDN1) following EVs exposure was decreased by HAc40 and c40 treatment, indicating a protective effect on the epidermal barrier's function. These findings demonstrate how Hac40 and c40 may mitigate the harmful effects of S. aureus EVs. Further investigation is needed to elucidate the exact mechanisms underlying this interaction and explore the potential clinical utility of c40 and its mucopolysaccharide carrier conjugate HAc40 in managing atopic dermatitis.
Collapse
Affiliation(s)
- Giulio Petronio Petronio
- Department of Medicina e Scienze della Salute “V. Tiberio”, Università degli Studi del Molise, 86100 Campobasso, Italy (R.D.M.)
| | - Maria Di Naro
- Department of Drug and Health Sciences, Università degli Studi di Catania, 95125 Catania, Italy
| | - Noemi Venditti
- Department of Medicina e Scienze della Salute “V. Tiberio”, Università degli Studi del Molise, 86100 Campobasso, Italy (R.D.M.)
- UO Laboratorio Analisi, Responsible Research Hospital, 86100 Campobasso, Italy
| | - Antonio Guarnieri
- Department of Medicina e Scienze della Salute “V. Tiberio”, Università degli Studi del Molise, 86100 Campobasso, Italy (R.D.M.)
| | | | | | - Alessandro Medoro
- Department of Medicina e Scienze della Salute “V. Tiberio”, Università degli Studi del Molise, 86100 Campobasso, Italy (R.D.M.)
| | - Emanuele Foderà
- Department of Medicina e Scienze della Salute “V. Tiberio”, Università degli Studi del Molise, 86100 Campobasso, Italy (R.D.M.)
| | - Daniela Passarella
- Department of Medicina e Scienze della Salute “V. Tiberio”, Università degli Studi del Molise, 86100 Campobasso, Italy (R.D.M.)
| | - Daria Nicolosi
- Department of Drug and Health Sciences, Università degli Studi di Catania, 95125 Catania, Italy
| | - Roberto Di Marco
- Department of Medicina e Scienze della Salute “V. Tiberio”, Università degli Studi del Molise, 86100 Campobasso, Italy (R.D.M.)
| |
Collapse
|
11
|
Galvan A, Pellicciari C, Calderan L. Recreating Human Skin In Vitro: Should the Microbiota Be Taken into Account? Int J Mol Sci 2024; 25:1165. [PMID: 38256238 PMCID: PMC10816982 DOI: 10.3390/ijms25021165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 01/12/2024] [Accepted: 01/16/2024] [Indexed: 01/24/2024] Open
Abstract
Skin plays crucial roles in the human body: besides protecting the organism from external threats, it acts as a thermal regulator, is responsible for the sense of touch, hosts microbial communities (the skin microbiota) involved in preventing the invasion of foreign pathogens, contains immunocompetent cells that maintain a healthy immunogenic/tolerogenic balance, and is a suitable route for drug administration. In the skin, four defense levels can be identified: besides the physical, chemical, and immune barriers that are inherent to the tissue, the skin microbiota (i.e., the numerous microorganisms living on the skin surface) provides an additional barrier. Studying the skin barrier function or the effects of drugs or cosmetic agents on human skin is a difficult task since snapshot evidence can only be obtained using bioptic samples where dynamic processes cannot properly be followed. To overcome these limitations, many different in vitro models of human skin have been developed that are characterized by diverse levels of complexity in terms of chemical, structural, and cellular composition. The aim of this review is to summarize and discuss the advantages and disadvantages of the different human skin models so far available and to underline how the insertion of a proper microbiota would positively impact an in vitro human skin model in an attempt to better mimic conditions in vivo.
Collapse
Affiliation(s)
- Andrea Galvan
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, 37134 Verona, Italy; (A.G.); (L.C.)
| | - Carlo Pellicciari
- Department of Biology and Biotechnology, University of Pavia, Via A. Ferrata 9, 27100 Pavia, Italy
| | - Laura Calderan
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, 37134 Verona, Italy; (A.G.); (L.C.)
| |
Collapse
|
12
|
Le Bras C, Rault L, Jacquet N, Daniel N, Chuat V, Valence F, Bellanger A, Bousarghin L, Blat S, Le Loir Y, Le Huërou-Luron I, Even S. Two human milk-like synthetic bacterial communities displayed contrasted impacts on barrier and immune responses in an intestinal quadricellular model. ISME COMMUNICATIONS 2024; 4:ycad019. [PMID: 38415201 PMCID: PMC10897888 DOI: 10.1093/ismeco/ycad019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/09/2023] [Revised: 12/22/2023] [Accepted: 12/26/2023] [Indexed: 02/29/2024]
Abstract
The human milk (HM) microbiota, a highly diverse microbial ecosystem, is thought to contribute to the health benefits associated with breast-feeding, notably through its impact on infant gut microbiota. Our objective was to further explore the role of HM bacteria on gut homeostasis through a "disassembly/reassembly" strategy. HM strains covering the diversity of HM cultivable microbiota were first characterized individually and then assembled in synthetic bacterial communities (SynComs) using two human cellular models, peripheral blood mononuclear cells and a quadricellular model mimicking intestinal epithelium. Selected HM bacteria displayed a large range of immunomodulatory properties and had variable effects on epithelial barrier, allowing their classification in functional groups. This multispecies characterization of HM bacteria showed no clear association between taxonomy and HM bacteria impacts on epithelial immune and barrier functions, revealing the entirety and complexity of HM bacteria potential. More importantly, the assembly of HM strains into two SynComs of similar taxonomic composition but with strains exhibiting distinct individual properties, resulted in contrasting impacts on the epithelium. These impacts of SynComs partially diverged from the predicted ones based on individual bacteria. Overall, our results indicate that the functional properties of the HM bacterial community rather than the taxonomic composition itself could play a crucial role in intestinal homeostasis of infants.
Collapse
Affiliation(s)
- Charles Le Bras
- STLO, INRAE, Institut Agro, Rennes, 35042, France
- Institut NuMeCan, INRAE, INSERM, Univ Rennes, Rennes-Saint Gilles, 35590, France
| | - Lucie Rault
- STLO, INRAE, Institut Agro, Rennes, 35042, France
| | | | | | | | | | | | - Latifa Bousarghin
- Institut NuMeCan, INRAE, INSERM, Univ Rennes, Rennes-Saint Gilles, 35590, France
| | - Sophie Blat
- Institut NuMeCan, INRAE, INSERM, Univ Rennes, Rennes-Saint Gilles, 35590, France
| | - Yves Le Loir
- STLO, INRAE, Institut Agro, Rennes, 35042, France
| | | | - Sergine Even
- STLO, INRAE, Institut Agro, Rennes, 35042, France
| |
Collapse
|
13
|
Szabó K, Bolla BS, Erdei L, Balogh F, Kemény L. Are the Cutaneous Microbiota a Guardian of the Skin's Physical Barrier? The Intricate Relationship between Skin Microbes and Barrier Integrity. Int J Mol Sci 2023; 24:15962. [PMID: 37958945 PMCID: PMC10647730 DOI: 10.3390/ijms242115962] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 10/31/2023] [Accepted: 11/01/2023] [Indexed: 11/15/2023] Open
Abstract
The skin is a tightly regulated, balanced interface that maintains our integrity through a complex barrier comprising physical or mechanical, chemical, microbiological, and immunological components. The skin's microbiota affect various properties, one of which is the establishment and maintenance of the physical barrier. This is achieved by influencing multiple processes, including keratinocyte differentiation, stratum corneum formation, and regulation of intercellular contacts. In this review, we summarize the potential contribution of Cutibacterium acnes to these events and outline the contribution of bacterially induced barrier defects to the pathogenesis of acne vulgaris. With the combined effects of a Westernized lifestyle, microbial dysbiosis, epithelial barrier defects, and inflammation, the development of acne is very similar to that of several other multifactorial diseases of barrier organs (e.g., inflammatory bowel disease, celiac disease, asthma, atopic dermatitis, and chronic rhinosinusitis). Therefore, the management of acne requires a complex approach, which should be taken into account when designing novel treatments that address not only the inflammatory and microbial components but also the maintenance and strengthening of the cutaneous physical barrier.
Collapse
Affiliation(s)
- Kornélia Szabó
- HUN-REN-SZTE Dermatological Research Group, 6720 Szeged, Hungary
- Department of Dermatology and Allergology, Albert Szent-Györgyi Medical School, University of Szeged, 6720 Szeged, Hungary; (B.S.B.)
- HCEMM-USZ Skin Research Group, 6720 Szeged, Hungary
| | - Beáta Szilvia Bolla
- Department of Dermatology and Allergology, Albert Szent-Györgyi Medical School, University of Szeged, 6720 Szeged, Hungary; (B.S.B.)
- HCEMM-USZ Skin Research Group, 6720 Szeged, Hungary
| | - Lilla Erdei
- Department of Dermatology and Allergology, Albert Szent-Györgyi Medical School, University of Szeged, 6720 Szeged, Hungary; (B.S.B.)
- HCEMM-USZ Skin Research Group, 6720 Szeged, Hungary
| | - Fanni Balogh
- HUN-REN-SZTE Dermatological Research Group, 6720 Szeged, Hungary
- Department of Dermatology and Allergology, Albert Szent-Györgyi Medical School, University of Szeged, 6720 Szeged, Hungary; (B.S.B.)
| | - Lajos Kemény
- HUN-REN-SZTE Dermatological Research Group, 6720 Szeged, Hungary
- Department of Dermatology and Allergology, Albert Szent-Györgyi Medical School, University of Szeged, 6720 Szeged, Hungary; (B.S.B.)
- HCEMM-USZ Skin Research Group, 6720 Szeged, Hungary
| |
Collapse
|
14
|
Park HY, Kweon DK, Kim JK. Upregulation of tight junction-related proteins by hyaluronic acid in human HaCaT keratinocytes. BIOACTIVE CARBOHYDRATES AND DIETARY FIBRE 2023; 30:100374. [DOI: 10.1016/j.bcdf.2023.100374] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
15
|
Moran MC, Brewer MG, Schlievert PM, Beck LA. S. aureus virulence factors decrease epithelial barrier function and increase susceptibility to viral infection. Microbiol Spectr 2023; 11:e0168423. [PMID: 37737609 PMCID: PMC10581065 DOI: 10.1128/spectrum.01684-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Accepted: 07/03/2023] [Indexed: 09/23/2023] Open
Abstract
Individuals with atopic dermatitis (AD) are highly colonized by Staphylococcus aureus and are more susceptible to severe viral complications. We hypothesized that S. aureus secreted virulence factors may alter keratinocyte biology to enhance viral susceptibility through disruption of the skin barrier, impaired keratinocyte differentiation, and/or inflammation. To address this hypothesis, human keratinocytes were exposed to conditioned media from multiple S. aureus strains that vary in virulence factor production (USA300, HG003, and RN4220) or select purified virulence factors. We have identified the S. aureus enterotoxin-like superantigen SElQ, as a virulence factor of interest, since it is highly produced by USA300 and was detected on the skin of 53% of AD subjects (n = 72) in a study conducted by our group. Treatment with USA300 conditioned media or purified SElQ resulted in a significant increase in keratinocyte susceptibility to infection with vaccinia virus, and also significantly decreased barrier function. Importantly, we have previously demonstrated that keratinocyte differentiation influences susceptibility to viral infection, and our qPCR observations indicated that USA300 S. aureus and SElQ alter differentiation in keratinocytes. CRISPR/Cas9 was used to knock out CD40, a potential enterotoxin receptor on epithelial cells. We found that CD40 expression on keratinocytes was not completely necessary for SElQ-mediated responses, as measured by proinflammatory cytokine expression and barrier function. Together, these findings support that select S. aureus virulence factors, particularly SElQ, enhance the susceptibility of epidermal cells to viral infection, which may contribute to the increased cutaneous infections observed in individuals with AD. IMPORTANCE Staphylococcus aureus skin colonization and infection are frequently observed in individuals with atopic dermatitis. Many S. aureus strains belong to the clonal group USA300, and these strains produce superantigens including the staphylococcal enterotoxin-like Q (SElQ). Our studies highlight that SElQ may play a key role by altering keratinocyte differentiation and reducing barrier function; collectively, this may explain the AD-specific enhanced infection risk to cutaneous viruses. It is unclear what receptor mediates SElQ's effects on keratinocytes. We have shown that one putative surface receptor, CD40, was not critical for its effects on proinflammatory cytokine production or barrier function.
Collapse
Affiliation(s)
- Mary C. Moran
- Department of Dermatology, University of Rochester Medical Center, Rochester, New York, USA
- Department of Microbiology & Immunology, University of Rochester Medical Center, Rochester, New York, USA
| | - Matthew G. Brewer
- Department of Dermatology, University of Rochester Medical Center, Rochester, New York, USA
| | | | - Lisa A. Beck
- Department of Dermatology, University of Rochester Medical Center, Rochester, New York, USA
| |
Collapse
|
16
|
Landemaine L, Da Costa G, Fissier E, Francis C, Morand S, Verbeke J, Michel ML, Briandet R, Sokol H, Gueniche A, Bernard D, Chatel JM, Aguilar L, Langella P, Clavaud C, Richard ML. Staphylococcus epidermidis isolates from atopic or healthy skin have opposite effect on skin cells: potential implication of the AHR pathway modulation. Front Immunol 2023; 14:1098160. [PMID: 37304256 PMCID: PMC10250813 DOI: 10.3389/fimmu.2023.1098160] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Accepted: 05/04/2023] [Indexed: 06/13/2023] Open
Abstract
Introduction Staphylococcus epidermidis is a commensal bacterium ubiquitously present on human skin. This species is considered as a key member of the healthy skin microbiota, involved in the defense against pathogens, modulating the immune system, and involved in wound repair. Simultaneously, S. epidermidis is the second cause of nosocomial infections and an overgrowth of S. epidermidis has been described in skin disorders such as atopic dermatitis. Diverse isolates of S. epidermidis co-exist on the skin. Elucidating the genetic and phenotypic specificities of these species in skin health and disease is key to better understand their role in various skin conditions. Additionally, the exact mechanisms by which commensals interact with host cells is partially understood. We hypothesized that S. epidermidis isolates identified from different skin origins could play distinct roles on skin differentiation and that these effects could be mediated by the aryl hydrocarbon receptor (AhR) pathway. Methods For this purpose, a library of 12 strains originated from healthy skin (non-hyperseborrheic (NH) and hyperseborrheic (H) skin types) and disease skin (atopic (AD) skin type) was characterized at the genomic and phenotypic levels. Results and discussion Here we showed that strains from atopic lesional skin alter the epidermis structure of a 3D reconstructed skin model whereas strains from NH healthy skin do not. All strains from NH healthy skin induced AhR/OVOL1 path and produced high quantities of indole metabolites in co-culture with NHEK; especially indole-3-aldehyde (IAld) and indole-3-lactic acid (ILA); while AD strains did not induce AhR/OVOL1 path but its inhibitor STAT6 and produced the lowest levels of indoles as compared to the other strains. As a consequence, strains from AD skin altered the differentiation markers FLG and DSG1. The results presented here, on a library of 12 strains, showed that S. epidermidis originated from NH healthy skin and atopic skin have opposite effects on the epidermal cohesion and structure and that these differences could be linked to their capacity to produce metabolites, which in turn could activate AHR pathway. Our results on a specific library of strains provide new insights into how S. epidermidis may interact with the skin to promote health or disease.
Collapse
Affiliation(s)
- Leslie Landemaine
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, Jouy-en-Josas, France
- L’Oréal Research and Innovation, Aulnay-sous-Bois, France
| | - Gregory Da Costa
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, Jouy-en-Josas, France
- Paris Center for Microbiome Medicine (PaCeMM), Fédération Hospitalo-Universitaire, Paris, France
| | - Elsa Fissier
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, Jouy-en-Josas, France
- Paris Center for Microbiome Medicine (PaCeMM), Fédération Hospitalo-Universitaire, Paris, France
| | - Carine Francis
- L’Oréal Research and Innovation, Aulnay-sous-Bois, France
| | | | | | - Marie-Laure Michel
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, Jouy-en-Josas, France
- Paris Center for Microbiome Medicine (PaCeMM), Fédération Hospitalo-Universitaire, Paris, France
| | - Romain Briandet
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, Jouy-en-Josas, France
| | - Harry Sokol
- Paris Center for Microbiome Medicine (PaCeMM), Fédération Hospitalo-Universitaire, Paris, France
- Sorbonne Université, INSERM UMRS-938, Centre de Recherche Saint-Antoine, Assistance Publique - Hôpitaux de Paris (AP-HP), Paris, France
| | | | | | - Jean-Marc Chatel
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, Jouy-en-Josas, France
- Paris Center for Microbiome Medicine (PaCeMM), Fédération Hospitalo-Universitaire, Paris, France
| | - Luc Aguilar
- L’Oréal Research and Innovation, Aulnay-sous-Bois, France
| | - Philippe Langella
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, Jouy-en-Josas, France
- Paris Center for Microbiome Medicine (PaCeMM), Fédération Hospitalo-Universitaire, Paris, France
| | - Cecile Clavaud
- L’Oréal Research and Innovation, Aulnay-sous-Bois, France
| | - Mathias L. Richard
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, Jouy-en-Josas, France
- Paris Center for Microbiome Medicine (PaCeMM), Fédération Hospitalo-Universitaire, Paris, France
| |
Collapse
|
17
|
Magnifico I, Perna A, Cutuli MA, Medoro A, Pietrangelo L, Guarnieri A, Foderà E, Passarella D, Venditti N, Vergalito F, Petronio Petronio G, Di Marco R. A Wall Fragment of Cutibacterium acnes Preserves Junctional Integrity Altered by Staphylococcus aureus in an Ex Vivo Porcine Skin Model. Pharmaceutics 2023; 15:pharmaceutics15041224. [PMID: 37111709 PMCID: PMC10145065 DOI: 10.3390/pharmaceutics15041224] [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: 02/27/2023] [Revised: 04/07/2023] [Accepted: 04/07/2023] [Indexed: 04/29/2023] Open
Abstract
(1) Background alteration of the skin microbiota, dysbiosis, causes skin barrier impairment resulting in disease development. Staphylococcus aureus, the main pathogen associated with dysbiosis, secretes several virulence factors, including α-toxin that damages tight junctions and compromises the integrity of the skin barrier. The use of members of the resident microbiota to restore the skin barrier, bacteriotherapy, represents a safe treatment for skin conditions among innovative options. The aim of this study is the evaluation of a wall fragment derived from a patented strain of Cutibacterium acnes DSM28251 (c40) alone and conjugated to a mucopolysaccharide carrier (HAc40) in counteracting S. aureus pathogenic action on two tight junction proteins (Claudin-1 and ZO-1) in an ex vivo porcine skin infection model. Methods: skin biopsies were infected with live S. aureus strains ATCC29213 and DSM20491. Tissue was pre-incubated or co-incubated with c40 and HAc40. (3) Results: c40 and HAc40 prevent and counteract Claudin-1 and Zo-1 damage (4) Conclusions: c40 and the functional ingredient HAc40 represent a potential non-pharmacological treatment of skin diseases associated with cutaneous dysbiosis of S. aureus. These findings offer numerous avenues for new research.
Collapse
Affiliation(s)
- Irene Magnifico
- Department of Medicine and Health Science "V. Tiberio", Università degli Studi del Molise, 8600 Campobasso, Italy
| | - Angelica Perna
- Department of Medicine and Health Science "V. Tiberio", Università degli Studi del Molise, 8600 Campobasso, Italy
| | - Marco Alfio Cutuli
- Department of Medicine and Health Science "V. Tiberio", Università degli Studi del Molise, 8600 Campobasso, Italy
| | - Alessando Medoro
- Department of Medicine and Health Science "V. Tiberio", Università degli Studi del Molise, 8600 Campobasso, Italy
| | - Laura Pietrangelo
- Department of Medicine and Health Science "V. Tiberio", Università degli Studi del Molise, 8600 Campobasso, Italy
| | - Antonio Guarnieri
- Department of Medicine and Health Science "V. Tiberio", Università degli Studi del Molise, 8600 Campobasso, Italy
| | - Emanuele Foderà
- Department of Medicine and Health Science "V. Tiberio", Università degli Studi del Molise, 8600 Campobasso, Italy
| | - Daniela Passarella
- Department of Medicine and Health Science "V. Tiberio", Università degli Studi del Molise, 8600 Campobasso, Italy
| | - Noemi Venditti
- Department of Medicine and Health Science "V. Tiberio", Università degli Studi del Molise, 8600 Campobasso, Italy
| | - Franca Vergalito
- Department of Agricultural, Environmental and Food Sciences (DiAAA), Università degli Studi del Molise, 8600 Campobasso, Italy
| | - Giulio Petronio Petronio
- Department of Medicine and Health Science "V. Tiberio", Università degli Studi del Molise, 8600 Campobasso, Italy
| | - Roberto Di Marco
- Department of Medicine and Health Science "V. Tiberio", Università degli Studi del Molise, 8600 Campobasso, Italy
| |
Collapse
|
18
|
Kingsley C, Kourtidis A. Critical roles of adherens junctions in diseases of the oral mucosa. Tissue Barriers 2023; 11:2084320. [PMID: 35659464 PMCID: PMC10161952 DOI: 10.1080/21688370.2022.2084320] [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: 04/07/2022] [Revised: 05/18/2022] [Accepted: 05/26/2022] [Indexed: 10/18/2022] Open
Abstract
The oral cavity is directly exposed to a variety of environmental stimuli and contains a diverse microbiome that continuously interacts with the oral epithelium. Therefore, establishment and maintenance of the barrier function of the oral mucosa is of paramount importance for its function and for the body's overall health. The adherens junction is a cell-cell adhesion complex that is essential for epithelial barrier function. Although a considerable body of work has associated barrier disruption with oral diseases, the molecular underpinnings of these associations have not been equally investigated. This is critical, since adherens junction components also possess significant signaling roles in the cell, in addition to their architectural ones. Here, we summarize current knowledge involving adherens junction components in oral pathologies, such as cancer and oral pathogen-related diseases, while we also discuss gaps in the knowledge and opportunities for future investigation of the relationship between adherens junctions and oral diseases.
Collapse
Affiliation(s)
- Christina Kingsley
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC, USA
| | - Antonis Kourtidis
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC, USA
| |
Collapse
|
19
|
Park HY, Kweon DK, Kim JK. Molecular weight-dependent hyaluronic acid permeability and tight junction modulation in human buccal TR146 cell monolayers. Int J Biol Macromol 2023; 227:182-192. [PMID: 36529213 DOI: 10.1016/j.ijbiomac.2022.12.106] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 11/22/2022] [Accepted: 12/11/2022] [Indexed: 12/23/2022]
Abstract
The oral route is considered an attractive method for drug delivery, as it avoids the hepatic and intestinal first-pass metabolism processes. Hyaluronic acid (HA) beneficial effects to the human body include anti-aging and wound healing but its effects on oral barrier integrity and mechanical function have not yet been investigated. In this study, we analyzed oral barrier integrity and the paracellular pathway of HA transportation in TR146 cell monolayers during and after permeation and using low molecular weight HA (LMW-HA, <100 kDa) and high molecular weight HA (HMW-HA, >500 kDa). Cytotoxicity assays in TR146 cells revealed that neither LMW-HA or HMW-HA altered cell viability at concentrations <0.5 % during 24 h of treatment. HA-treated TR146 cell monolayers showed enhanced oral barrier integrity and reduced apparent permeability of fluorescein. Moreover, HA significantly increased tight junction (TJ)-related genes expression, including ZO-2, marvelD3, cingulin, claudin-1, claudin-3, and claudin-4 expression. Overall, the results of the present study indicate that HA can permeate across the oral barrier and enhance oral barrier function via the upregulated expression of TJ-related genes.
Collapse
Affiliation(s)
- Ha-Young Park
- Advanced Radiation Technology Institute, Korea Atomic Energy Research Institute, Jeongeup 56212, Republic of Korea.
| | - Dong-Keon Kweon
- Jinwoo Bio Co., Ltd., Giheung-gu, Yongin-si 17111, Republic of Korea
| | - Jae-Kyung Kim
- Advanced Radiation Technology Institute, Korea Atomic Energy Research Institute, Jeongeup 56212, Republic of Korea.
| |
Collapse
|
20
|
Francis D, Bhairaddy A, Joy A, Hari GV, Francis A. Secretory proteins in the orchestration of microbial virulence: The curious case of Staphylococcus aureus. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2023; 133:271-350. [PMID: 36707204 DOI: 10.1016/bs.apcsb.2022.10.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Microbial virulence showcases an excellent model for adaptive changes that enable an organism to survive and proliferate in a hostile environment and exploit host resources to its own benefit. In Staphylococcus aureus, an opportunistic pathogen of the human host, known for the diversity of the disease conditions it inflicts and the rapid evolution of antibiotic resistance, virulence is a consequence of having a highly plastic genome that is amenable to quick reprogramming and the ability to express a diverse arsenal of virulence factors. Virulence factors that are secreted to the host milieu effectively manipulate the host conditions to favor bacterial survival and growth. They assist in colonization, nutrient acquisition, immune evasion, and systemic spread. The structural and functional characteristics of the secreted virulence proteins have been shaped to assist S. aureus in thriving and disseminating effectively within the host environment and exploiting the host resources to its best benefit. With the aim of highlighting the importance of secreted virulence proteins in bacterial virulence, the present chapter provides a comprehensive account of the role of the major secreted proteins of S. aureus in orchestrating its virulence in the human host.
Collapse
Affiliation(s)
- Dileep Francis
- Department of Life Sciences, Kristu Jayanti College, Autonomous, Bengaluru, Karnataka, India.
| | - Anusha Bhairaddy
- Department of Life Sciences, Kristu Jayanti College, Autonomous, Bengaluru, Karnataka, India
| | - Atheene Joy
- Department of Life Sciences, Kristu Jayanti College, Autonomous, Bengaluru, Karnataka, India
| | | | - Ashik Francis
- Rajiv Gandhi Centre for Biotechnology, Trivandrum, Kerala, India
| |
Collapse
|
21
|
Herpes Simplex Virus 1 Can Bypass Impaired Epidermal Barriers upon Ex Vivo Infection of Skin from Atopic Dermatitis Patients. J Virol 2022; 96:e0086422. [PMID: 35969080 PMCID: PMC9472615 DOI: 10.1128/jvi.00864-22] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
To infect its human host, herpes simplex virus 1 (HSV-1) must overcome the protective barriers of skin and mucosa. Here, we addressed whether pathological skin conditions can facilitate viral entry via the skin surface and used ex vivo infection studies to explore viral invasion in atopic dermatitis (AD) skin characterized by disturbed barrier functions. Our focus was on the visualization of the onset of infection in single cells to determine the primary entry portals in the epidermis. After ex vivo infection of lesional AD skin, we observed infected cells in suprabasal layers indicating successful invasion in the epidermis via the skin surface which was never detected in control skin where only sample edges allowed viral access. The redistribution of filaggrin, loricrin, and tight-junction components in the lesional skin samples suggested multiple defective mechanical barriers. To dissect the parameters that contribute to HSV-1 invasion, we induced an AD-like phenotype by adding the Th2 cytokines interleukin 4 (IL-4) and IL-13 to healthy human skin samples. Strikingly, we detected infected cells in the epidermis, implying that the IL-4/IL-13-driven inflammation is sufficient to induce modifications allowing HSV-1 to penetrate the skin surface. In summary, not only did lesional AD skin facilitate HSV-1 penetration but IL-4/IL-13 responses alone allowed virus invasion. Our results suggest that the defective epidermal barriers of AD skin and the inflammation-induced altered barriers in healthy skin can make receptors accessible for HSV-1. IMPORTANCE Herpes simplex virus 1 (HSV-1) can target skin to establish primary infection in the epithelium. While the human skin provides effective barriers against viral invasion under healthy conditions, a prominent example of successful invasion is the disseminated HSV-1 infection in the skin of atopic dermatitis (AD) patients. AD is characterized by impaired epidermal barrier functions, chronic inflammation, and dysbiosis of skin microbiota. We addressed the initial invasion process of HSV-1 in atopic dermatitis skin to understand whether the physical barrier functions are sufficiently disturbed to allow the virus to invade skin and reach its receptors on skin cells. Our results demonstrate that HSV-1 can indeed penetrate and initiate infection in atopic dermatitis skin. Since treatment of skin with IL-4 and IL-13 already resulted in successful invasion, we assume that inflammation-induced barrier defects play an important role for the facilitated access of HSV-1 to its target cells.
Collapse
|
22
|
Oral and external intervention on the crosstalk between microbial barrier and skin via foodborne functional component. J Funct Foods 2022. [DOI: 10.1016/j.jff.2022.105075] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
|
23
|
Beck LA, Cork MJ, Amagai M, De Benedetto A, Kabashima K, Hamilton JD, Rossi AB. Type 2 Inflammation Contributes to Skin Barrier Dysfunction in Atopic Dermatitis. JID INNOVATIONS 2022; 2:100131. [PMID: 36059592 PMCID: PMC9428921 DOI: 10.1016/j.xjidi.2022.100131] [Citation(s) in RCA: 115] [Impact Index Per Article: 38.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 01/04/2022] [Accepted: 01/06/2022] [Indexed: 01/02/2023] Open
Abstract
Skin barrier dysfunction, a defining feature of atopic dermatitis (AD), arises from multiple interacting systems. In AD, skin inflammation is caused by host-environment interactions involving keratinocytes as well as tissue-resident immune cells such as type 2 innate lymphoid cells, basophils, mast cells, and T helper type 2 cells, which produce type 2 cytokines, including IL-4, IL-5, IL-13, and IL-31. Type 2 inflammation broadly impacts the expression of genes relevant for barrier function, such as intracellular structural proteins, extracellular lipids, and junctional proteins, and enhances Staphylococcus aureus skin colonization. Systemic anti‒type 2 inflammation therapies may improve dysfunctional skin barrier in AD.
Collapse
Key Words
- AD, atopic dermatitis
- AMP, antimicrobial peptide
- CLDN, claudin
- FFA, free fatty acid
- ILC2, type 2 innate lymphoid cell
- Jaki, Jak inhibitor
- K, keratin
- KC, keratinocyte
- MMP, matrix metalloproteinase
- NMF, natural moisturizing factor
- PAR, protease-activated receptor
- PDE-4, phosphodiesterase-4
- SC, stratum corneum
- SG, stratum granulosum
- TCI, topical calcineurin inhibitor
- TCS, topical corticosteroid
- TEWL, transepidermal water loss
- TJ, tight junction
- TLR, toll-like receptor
- TNF-α, tumor necrosis factor alpha
- TYK, tyrosine kinase
- Th, T helper
- ZO, zona occludens
- hBD, human β-defensin
Collapse
Affiliation(s)
- Lisa A. Beck
- Department of Dermatology, University of Rochester Medical Center, Rochester, New York, USA,Correspondence: Lisa A. Beck, Department of Dermatology, University of Rochester Medical Center, 601 Elmwood Ave, Box 697, Rochester, New York 14642, USA.
| | - Michael J. Cork
- Sheffield Dermatology Research, Department of Infection, Immunity and Cardiovascular Disease (IICD), The University of Sheffield, The Medical School, Sheffield, United Kingdom
| | - Masayuki Amagai
- Department of Dermatology, Keio University School of Medicine, Tokyo, Japan,Laboratory for Skin Homeostasis, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Anna De Benedetto
- Department of Dermatology, University of Rochester Medical Center, Rochester, New York, USA
| | - Kenji Kabashima
- Department of Dermatology, Kyoto University Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | | | | |
Collapse
|
24
|
Micronutrient Improvement of Epithelial Barrier Function in Various Disease States: A Case for Adjuvant Therapy. Int J Mol Sci 2022; 23:ijms23062995. [PMID: 35328419 PMCID: PMC8951934 DOI: 10.3390/ijms23062995] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 03/01/2022] [Indexed: 02/04/2023] Open
Abstract
The published literature makes a very strong case that a wide range of disease morbidity associates with and may in part be due to epithelial barrier leak. An equally large body of published literature substantiates that a diverse group of micronutrients can reduce barrier leak across a wide array of epithelial tissue types, stemming from both cell culture as well as animal and human tissue models. Conversely, micronutrient deficiencies can exacerbate both barrier leak and morbidity. Focusing on zinc, Vitamin A and Vitamin D, this review shows that at concentrations above RDA levels but well below toxicity limits, these micronutrients can induce cell- and tissue-specific molecular-level changes in tight junctional complexes (and by other mechanisms) that reduce barrier leak. An opportunity now exists in critical care—but also medical prophylactic and therapeutic care in general—to consider implementation of select micronutrients at elevated dosages as adjuvant therapeutics in a variety of disease management. This consideration is particularly pointed amidst the COVID-19 pandemic.
Collapse
|
25
|
Herman KE, Yoshida T, Hughson A, Grier A, Gill SR, Beck LA, Fowell DJ. IL-17-Dependent Dysregulated Cutaneous Immune Homeostasis in the Absence of the Wiskott-Aldrich Syndrome Protein. Front Immunol 2022; 13:817427. [PMID: 35265075 PMCID: PMC8900519 DOI: 10.3389/fimmu.2022.817427] [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: 11/18/2021] [Accepted: 01/17/2022] [Indexed: 11/13/2022] Open
Abstract
Wiskott-Aldrich Syndrome (WAS) is characterized by recurrent infections, thrombocytopenia, and eczema. Here, we show that WASp-deficient mice on a BALB/c background have dysregulated cutaneous immune homeostasis with increased leukocyte accumulation in the skin, 1 week after birth. Increased cutaneous inflammation was associated with epithelial abnormalities, namely, altered keratinization, abnormal epidermal tight junctional morphology and increased trans-epidermal water loss; consistent with epidermal barrier dysfunction. Immune and physical barrier disruption was accompanied by progressive skin dysbiosis, highlighting the functional significance of the disrupted cutaneous homeostasis. Interestingly, the dysregulated immunity in the skin preceded the systemic elevation in IgE and lymphocytic infiltration of the colonic lamina propria associated with WASp deficiency. Mechanistically, the enhanced immune cell accumulation in the skin was lymphocyte dependent. Elevated levels of both Type 2 (IL-4, IL-5) and Type 17 (IL-17, IL-22, IL-23) cytokines were present in the skin, as well as the 'itch' factor IL-31. Unexpectedly, the canonical WAS-associated cytokine IL-4 did not play a role in the immune dysfunction. Instead, IL-17 was critical for skin immune infiltration and elevation of both Type 2 and Type 17 cytokines. Our findings reveal a previously unrecognized IL-17-dependent breakdown in immune homeostasis and cutaneous barrier integrity in the absence of WASp, targeting of which may provide new therapeutic possibilities for the treatment of skin pathologies in WAS patients.
Collapse
Affiliation(s)
- Katherine E. Herman
- David H. Smith Center for Vaccine Biology and Immunology, Aab Institute of Biomedical Sciences, University of Rochester Medical Center, Rochester, NY, United States
| | - Takeshi Yoshida
- Department of Dermatology, University of Rochester Medical Center, Rochester, NY, United States
| | - Angela Hughson
- David H. Smith Center for Vaccine Biology and Immunology, Aab Institute of Biomedical Sciences, University of Rochester Medical Center, Rochester, NY, United States
| | - Alex Grier
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY, United States
| | - Steven R. Gill
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY, United States
| | - Lisa A. Beck
- Department of Dermatology, University of Rochester Medical Center, Rochester, NY, United States
| | - Deborah J. Fowell
- David H. Smith Center for Vaccine Biology and Immunology, Aab Institute of Biomedical Sciences, University of Rochester Medical Center, Rochester, NY, United States,Department of Microbiology and Immunology, Cornell University, Ithaca, NY, United States,*Correspondence: Deborah J. Fowell,
| |
Collapse
|
26
|
Advances in Microbiome-Derived Solutions and Methodologies Are Founding a New Era in Skin Health and Care. Pathogens 2022; 11:pathogens11020121. [PMID: 35215065 PMCID: PMC8879973 DOI: 10.3390/pathogens11020121] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 01/11/2022] [Accepted: 01/12/2022] [Indexed: 12/04/2022] Open
Abstract
The microbiome, as a community of microorganisms and their structural elements, genomes, metabolites/signal molecules, has been shown to play an important role in human health, with significant beneficial applications for gut health. Skin microbiome has emerged as a new field with high potential to develop disruptive solutions to manage skin health and disease. Despite an incomplete toolbox for skin microbiome analyses, much progress has been made towards functional dissection of microbiomes and host-microbiome interactions. A standardized and robust investigation of the skin microbiome is necessary to provide accurate microbial information and set the base for a successful translation of innovations in the dermo-cosmetic field. This review provides an overview of how the landscape of skin microbiome research has evolved from method development (multi-omics/data-based analytical approaches) to the discovery and development of novel microbiome-derived ingredients. Moreover, it provides a summary of the latest findings on interactions between the microbiomes (gut and skin) and skin health/disease. Solutions derived from these two paths are used to develop novel microbiome-based ingredients or solutions acting on skin homeostasis are proposed. The most promising skin and gut-derived microbiome interventional strategies are presented, along with regulatory, safety, industrial, and technical challenges related to a successful translation of these microbiome-based concepts/technologies in the dermo-cosmetic industry.
Collapse
|
27
|
Lefèvre-Utile A, Braun C, Haftek M, Aubin F. Five Functional Aspects of the Epidermal Barrier. Int J Mol Sci 2021; 22:11676. [PMID: 34769105 PMCID: PMC8583944 DOI: 10.3390/ijms222111676] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 10/24/2021] [Accepted: 10/25/2021] [Indexed: 01/07/2023] Open
Abstract
The epidermis is a living, multilayered barrier with five functional levels, including a physical, a chemical, a microbial, a neuronal, and an immune level. Altogether, this complex organ contributes to protect the host from external aggression and to preserve its integrity. In this review, we focused on the different functional aspects.
Collapse
Affiliation(s)
- Alain Lefèvre-Utile
- Sce de Pédiatrie Générale et Urgence pédiatrique, Hôpital Jean Verdier, Assistance Publique Hôpitaux de Paris, 93140 Bondy, France;
- Unité 976 HIPI, Institut de Recherche Saint-Louis, Université de Paris, Inserm, 75010, Paris, France
| | - Camille Braun
- Centre international de Recherche en Infectiologie, Inserm U1111, Université Claude Bernard Lyon 1, 69007 Lyon, France;
- Sce de Pneumologie Pédiatrique et Allergie, Hôpital Femme Mère Enfant, 69500 Bron, France
| | - Marek Haftek
- CNRS UMR5305, Laboratory of Tissue Biology and Therapeutic Engineering, LBTI, Lyon1 University, 69100 Lyon, France;
| | - François Aubin
- Inserm U1098, Université de Franche Comté, 25000 Besançon, France
- Sce de Dermatologie, Centre Hospitalier Universitaire, 25000 Besançon, France
| |
Collapse
|
28
|
Tonti OR, Larson H, Lipp SN, Luetkemeyer CM, Makam M, Vargas D, Wilcox SM, Calve S. Tissue-specific parameters for the design of ECM-mimetic biomaterials. Acta Biomater 2021; 132:83-102. [PMID: 33878474 PMCID: PMC8434955 DOI: 10.1016/j.actbio.2021.04.017] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Revised: 03/18/2021] [Accepted: 04/08/2021] [Indexed: 02/06/2023]
Abstract
The extracellular matrix (ECM) is a complex network of biomolecules that mechanically and biochemically directs cell behavior and is crucial for maintaining tissue function and health. The heterogeneous organization and composition of the ECM varies within and between tissue types, directing mechanics, aiding in cell-cell communication, and facilitating tissue assembly and reassembly during development, injury and disease. As technologies like 3D printing rapidly advance, researchers are better able to recapitulate in vivo tissue properties in vitro; however, tissue-specific variations in ECM composition and organization are not given enough consideration. This is in part due to a lack of information regarding how the ECM of many tissues varies in both homeostatic and diseased states. To address this gap, we describe the components and organization of the ECM, and provide examples for different tissues at various states of disease. While many aspects of ECM biology remain unknown, our goal is to highlight the complexity of various tissues and inspire engineers to incorporate unique components of the native ECM into in vitro platform design and fabrication. Ultimately, we anticipate that the use of biomaterials that incorporate key tissue-specific ECM will lead to in vitro models that better emulate human pathologies. STATEMENT OF SIGNIFICANCE: Biomaterial development primarily emphasizes the engineering of new materials and therapies at the expense of identifying key parameters of the tissue that is being emulated. This can be partially attributed to the difficulty in defining the 3D composition, organization, and mechanics of the ECM within different tissues and how these material properties vary as a function of homeostasis and disease. In this review, we highlight a range of tissues throughout the body and describe how ECM content, cell diversity, and mechanical properties change in diseased tissues and influence cellular behavior. Accurately mimicking the tissue of interest in vitro by using ECM specific to the appropriate state of homeostasis or pathology in vivo will yield results more translatable to humans.
Collapse
Affiliation(s)
- Olivia R Tonti
- Paul M. Rady Department of Mechanical Engineering, University of Colorado - Boulder, 1111 Engineering Center, 427 UCB, Boulder, CO 80309, United States
| | - Hannah Larson
- Paul M. Rady Department of Mechanical Engineering, University of Colorado - Boulder, 1111 Engineering Center, 427 UCB, Boulder, CO 80309, United States
| | - Sarah N Lipp
- Paul M. Rady Department of Mechanical Engineering, University of Colorado - Boulder, 1111 Engineering Center, 427 UCB, Boulder, CO 80309, United States
| | - Callan M Luetkemeyer
- Paul M. Rady Department of Mechanical Engineering, University of Colorado - Boulder, 1111 Engineering Center, 427 UCB, Boulder, CO 80309, United States
| | - Megan Makam
- Paul M. Rady Department of Mechanical Engineering, University of Colorado - Boulder, 1111 Engineering Center, 427 UCB, Boulder, CO 80309, United States
| | - Diego Vargas
- Paul M. Rady Department of Mechanical Engineering, University of Colorado - Boulder, 1111 Engineering Center, 427 UCB, Boulder, CO 80309, United States
| | - Sean M Wilcox
- Paul M. Rady Department of Mechanical Engineering, University of Colorado - Boulder, 1111 Engineering Center, 427 UCB, Boulder, CO 80309, United States
| | - Sarah Calve
- Paul M. Rady Department of Mechanical Engineering, University of Colorado - Boulder, 1111 Engineering Center, 427 UCB, Boulder, CO 80309, United States.
| |
Collapse
|
29
|
Blicharz L, Rudnicka L, Czuwara J, Waśkiel-Burnat A, Goldust M, Olszewska M, Samochocki Z. The Influence of Microbiome Dysbiosis and Bacterial Biofilms on Epidermal Barrier Function in Atopic Dermatitis-An Update. Int J Mol Sci 2021; 22:ijms22168403. [PMID: 34445108 PMCID: PMC8395079 DOI: 10.3390/ijms22168403] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 07/31/2021] [Accepted: 08/02/2021] [Indexed: 12/13/2022] Open
Abstract
Atopic dermatitis (AD) is a common inflammatory dermatosis affecting up to 30% of children and 10% of adults worldwide. AD is primarily driven by an epidermal barrier defect which triggers immune dysregulation within the skin. According to recent research such phenomena are closely related to the microbial dysbiosis of the skin. There is growing evidence that cutaneous microbiota and bacterial biofilms negatively affect skin barrier function, contributing to the onset and exacerbation of AD. This review summarizes the latest data on the mechanisms leading to microbiome dysbiosis and biofilm formation in AD, and the influence of these phenomena on skin barrier function.
Collapse
Affiliation(s)
- Leszek Blicharz
- Department of Dermatology, Medical University of Warsaw, 02-008 Warsaw, Poland; (L.R.); (J.C.); (A.W.-B.); (M.O.); (Z.S.)
- Correspondence:
| | - Lidia Rudnicka
- Department of Dermatology, Medical University of Warsaw, 02-008 Warsaw, Poland; (L.R.); (J.C.); (A.W.-B.); (M.O.); (Z.S.)
| | - Joanna Czuwara
- Department of Dermatology, Medical University of Warsaw, 02-008 Warsaw, Poland; (L.R.); (J.C.); (A.W.-B.); (M.O.); (Z.S.)
| | - Anna Waśkiel-Burnat
- Department of Dermatology, Medical University of Warsaw, 02-008 Warsaw, Poland; (L.R.); (J.C.); (A.W.-B.); (M.O.); (Z.S.)
| | - Mohamad Goldust
- Department of Dermatology, University Medical Center of the Johannes Gutenberg University, 55131 Mainz, Germany;
| | - Małgorzata Olszewska
- Department of Dermatology, Medical University of Warsaw, 02-008 Warsaw, Poland; (L.R.); (J.C.); (A.W.-B.); (M.O.); (Z.S.)
| | - Zbigniew Samochocki
- Department of Dermatology, Medical University of Warsaw, 02-008 Warsaw, Poland; (L.R.); (J.C.); (A.W.-B.); (M.O.); (Z.S.)
| |
Collapse
|
30
|
Boxberger M, Cenizo V, Cassir N, La Scola B. Challenges in exploring and manipulating the human skin microbiome. MICROBIOME 2021; 9:125. [PMID: 34053468 PMCID: PMC8166136 DOI: 10.1186/s40168-021-01062-5] [Citation(s) in RCA: 116] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Accepted: 03/25/2021] [Indexed: 05/08/2023]
Abstract
The skin is the exterior interface of the human body with the environment. Despite its harsh physical landscape, the skin is colonized by diverse commensal microbes. In this review, we discuss recent insights into skin microbial populations, including their composition and role in health and disease and their modulation by intrinsic and extrinsic factors, with a focus on the pathobiological basis of skin aging. We also describe the most recent tools for investigating the skin microbiota composition and microbe-skin relationships and perspectives regarding the challenges of skin microbiome manipulation. Video abstract.
Collapse
Affiliation(s)
- Manon Boxberger
- IRD, AP-HM, MEPHI, Aix Marseille Université, Marseille, France
- IHU-Méditerranée Infection, 19-21 Boulevard Jean Moulin, 13385 Marseille Cedex 05, France
| | - Valérie Cenizo
- Groupe L’Occitane, R&D Department, Zone Industrielle Saint Maurice, 4100 Manosque, Alpes-de Haute-Provence France
| | - Nadim Cassir
- IRD, AP-HM, MEPHI, Aix Marseille Université, Marseille, France
- IHU-Méditerranée Infection, 19-21 Boulevard Jean Moulin, 13385 Marseille Cedex 05, France
| | - Bernard La Scola
- IRD, AP-HM, MEPHI, Aix Marseille Université, Marseille, France
- IHU-Méditerranée Infection, 19-21 Boulevard Jean Moulin, 13385 Marseille Cedex 05, France
- IRD, AP-HM, SSA, VITROME, Aix Marseille Université, Marseille, France
| |
Collapse
|
31
|
Ohshima H, Kurosumi M, Kanto H. New solution of beauty problem by Staphylococcus hominis: Relevance between skin microbiome and skin condition in healthy subject. Skin Res Technol 2021; 27:692-700. [PMID: 33511688 DOI: 10.1111/srt.13001] [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: 09/25/2020] [Revised: 11/27/2020] [Accepted: 12/24/2020] [Indexed: 11/28/2022]
Abstract
BACKGROUND/AIMS Recently, it was suggested that skin microbiome is related to some skin disease. The possibility of affecting the skin might be high, but there were few reports of the influence on the skin condition in healthy subjects. Our aim was to evaluate the relationship between skin condition and skin microbiome in healthy subjects. METHODS Experiment 1: 293 Japanese healthy women were divided into two groups, good skin properties and poor skin properties by 14 skin physiology parameter values on the cheek using noninvasive method. Differences of abundance of bacterial species on the cheek between the two groups were evaluated. Experiment 2: 11 Japanese healthy women were applied Staphylococcus hominis (S. hominis) on half-side of cheek for eight times in 1 month. Difference of change of physiology parameter values comparing to placebo side was evaluated. RESULTS Experiment 1: Multiple skin bacterial species were found to be significantly relevant in 14 physiology parameters. The abundance of S. hominis on the cheek with good skin properties group was significantly higher than poor skin properties group. Experiment 2: The application of S. hominis improved significantly the conspicuous pore number, melanin index, and the wrinkle count compared to placebo side. CONCLUSION We found many skin bacterial species that might improve the skin condition in healthy women. In particular, S. hominis might have the potential to improve multiple skin beauty problems.
Collapse
Affiliation(s)
- Hiroshi Ohshima
- POLA Laboratories, POLA CHEMICAL INDUSTRIES, INC., Yokohama, Japan
| | | | - Hiromi Kanto
- Department of Dermatology, School of Medicine, Toho University, Tokyo, Japan
| |
Collapse
|
32
|
Schmidt A, Liebelt G, Striesow J, Freund E, von Woedtke T, Wende K, Bekeschus S. The molecular and physiological consequences of cold plasma treatment in murine skin and its barrier function. Free Radic Biol Med 2020; 161:32-49. [PMID: 33011275 DOI: 10.1016/j.freeradbiomed.2020.09.026] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 09/21/2020] [Accepted: 09/28/2020] [Indexed: 12/12/2022]
Abstract
Cold plasma technology is an emerging tool facilitating the spatially controlled delivery of a multitude of reactive species (ROS) to the skin. While the therapeutic efficacy of plasma treatment has been observed in several types of diseases, the fundamental consequences of plasma-derived ROS on skin physiology remain unknown. We aimed to bridge this gap since the epidermal skin barrier and perfusion plays a vital role in health and disease by maintaining homeostasis and protecting from environmental damage. The intact skin of SKH1 mice was plasma-treated in vivo. Gene and protein expression was analyzed utilizing transcriptomics, qPCR, and Western blot. Immunofluorescence aided the analysis of percutaneous skin penetration of curcumin. Tissue oxygenation, perfusion, hemoglobin, and water index was investigated using hyperspectral imaging. Reversed-phase liquid-chromatography/mass spectrometry was performed for the identification of changes in the lipid composition and oxidation. Transcriptomic analysis of plasma-treated skin revealed modulation of genes involved in regulating the junctional network (tight, adherence, and gap junctions), which was confirmed using qPCR, Western blot, and immunofluorescence imaging. Plasma treatment increased the disaggregation of cells in the stratum corneum (SC) concomitant with increased tissue oxygenation, gap junctional intercellular communication, and penetration of the model drug curcumin into the SC preceded by altered oxidation of skin lipids and their composition in vivo. In summary, plasma-derived ROS modify the junctional network, which promoted tissue oxygenation, oxidation of SC-lipids, and restricted penetration of the model drug curcumin, implicating that plasma may provide a novel and sensitive tool of skin barrier regulation.
Collapse
Affiliation(s)
- Anke Schmidt
- Plasma Life Science and ZIK Plasmatis, Leibniz Institute for Plasma Science and Technology (INP), Felix-Hausdorff-Str. 2, 17489, Greifswald, Germany
| | - Grit Liebelt
- Plasma Life Science and ZIK Plasmatis, Leibniz Institute for Plasma Science and Technology (INP), Felix-Hausdorff-Str. 2, 17489, Greifswald, Germany
| | - Johanna Striesow
- Plasma Life Science and ZIK Plasmatis, Leibniz Institute for Plasma Science and Technology (INP), Felix-Hausdorff-Str. 2, 17489, Greifswald, Germany
| | - Eric Freund
- Plasma Life Science and ZIK Plasmatis, Leibniz Institute for Plasma Science and Technology (INP), Felix-Hausdorff-Str. 2, 17489, Greifswald, Germany; Department of General Visceral, Thoracic, and Vascular Surgery, Greifswald University Medical Center, Sauerbruchstr. DZ7, 17475, Greifswald, Germany
| | - Thomas von Woedtke
- Plasma Life Science and ZIK Plasmatis, Leibniz Institute for Plasma Science and Technology (INP), Felix-Hausdorff-Str. 2, 17489, Greifswald, Germany; Institute for Hygiene and Environmental Medicine, Greifswald University Medical Center, Sauerbruchstr., 17489, Greifswald, Germany
| | - Kristian Wende
- Plasma Life Science and ZIK Plasmatis, Leibniz Institute for Plasma Science and Technology (INP), Felix-Hausdorff-Str. 2, 17489, Greifswald, Germany
| | - Sander Bekeschus
- Plasma Life Science and ZIK Plasmatis, Leibniz Institute for Plasma Science and Technology (INP), Felix-Hausdorff-Str. 2, 17489, Greifswald, Germany.
| |
Collapse
|
33
|
Galliano MF, Bäsler K, Caruana A, Mias C, Bessou-Touya S, Brandner JM, Duplan H. Protective effect of Aquaphilus dolomiae extract-G1, ADE-G1, on tight junction barrier function in a Staphylococcus aureus-infected atopic dermatitis model. J Eur Acad Dermatol Venereol 2020; 34 Suppl 5:30-36. [PMID: 32870557 DOI: 10.1111/jdv.16516] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Accepted: 02/15/2020] [Indexed: 11/30/2022]
Abstract
BACKGROUND Atopic dermatitis (AD) is a common skin disease characterized by recurrent pruritic inflammatory skin lesions and defects of the skin barrier. Bacterial infection with Staphylococcus aureus contributes to increased severity of AD by compromising the barrier further. A microorganism component of Avène Thermal Spring Water, Aquaphilus dolomiae, is thought to contribute to some of its beneficial effects to skin, eg AD alleviation. AIMS Here, we have investigated the effects of an extract of A. dolomiae, A. dolomiae extract-G1 (ADE-G1), on the structural barrier function of keratinocytes, tight junction (TJ) protein expression and the expression of several genes altered in AD patients. METHODS An epidermal cell culture model mimicking the AD environment and phenotype was used, in which S. aureus-infected cell cultures of normal human epidermal keratinocytes were exposed to a proinflammatory environment. Endpoints measured included the transepithelial electrical resistance (TER) and immunohistological staining of the epidermal TJ proteins, claudin and occludin. Additional analysis was made of several genes known to be differentially regulated in skin from AD patients (C-C motif chemokine ligand 20 (CCL20), interleukin-8 (IL-8), S100 calcium binding protein A7 (S100A7), defensin beta 4 (DEFB4) and filaggrin). RESULTS Aquaphilus dolomiae extract-G1 strongly increased TER in non-infected cells and provided protection against infection by overcoming the decrease in TER induced by the infection with S. aureus. In infected cells exposed to a pro-inflammatory environment - depicting AD-like conditions - TER protection by ADE-G1 was still observed. Gene expression analysis of infected and pro-inflammatory stimulated cells indicated that ADE-G1 modulated the inflammatory response (induced IL-8 and attenuated CCL20 expression), increased antimicrobial activities (induced DEFB4 and A100A7) and strengthened barrier function (restored filaggrin expression). CONCLUSIONS ADE-G1 reinforces barrier function and strongly protects TJ barrier disruption induced by bacterial infection and inflammation.
Collapse
Affiliation(s)
| | - K Bäsler
- Department of Dermatology and Venerology, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | - A Caruana
- Pierre Fabre Dermo-Cosmétique, Toulouse, France
| | - C Mias
- Pierre Fabre Dermo-Cosmétique, Toulouse, France
| | | | - J M Brandner
- Department of Dermatology and Venerology, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | - H Duplan
- Pierre Fabre Dermo-Cosmétique, Toulouse, France
| |
Collapse
|
34
|
Bolla BS, Erdei L, Urbán E, Burián K, Kemény L, Szabó K. Cutibacterium acnes regulates the epidermal barrier properties of HPV-KER human immortalized keratinocyte cultures. Sci Rep 2020; 10:12815. [PMID: 32733073 PMCID: PMC7393503 DOI: 10.1038/s41598-020-69677-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Accepted: 07/13/2020] [Indexed: 12/20/2022] Open
Abstract
Our skin provides a physical barrier to separate the internal part of our body from the environment. Maintenance of complex barrier functions is achieved through anatomical structures in the skin, the stratified squamous epithelium specialized junctional organelles, called tight junctions (TJs). Several members of our microbial communities are known to affect the differentiation state and function of the colonized organ. Whether and how interactions between skin cells and cutaneous microbes, including Cutibacterium acnes (C. acnes), modify the structure and/or function of our skin is currently only partly understood. Thus, in our studies, we investigated whether C. acnes may affect the epidermal barrier using in vitro model systems. Real-time cellular analysis showed that depending on the keratinocyte differentiation state, the applied C. acnes strains and their dose, the measured impedance values change, together with the expression of selected TJ proteins. These may reflect barrier alterations, which can be partially restored upon antibiotic–antimycotic treatment. Our findings suggest that C. acnes can actively modify the barrier properties of cultured keratinocytes, possibly through alteration of tight cell-to-cell contacts. Similar events may play important roles in our skin, in the maintenance of cutaneous homeostasis.
Collapse
Affiliation(s)
- Beáta Szilvia Bolla
- Department of Dermatology and Allergology, University of Szeged, Szeged, Hungary.,HCEMM-SZTE Skin Research Group, Szeged, Hungary
| | - Lilla Erdei
- Department of Dermatology and Allergology, University of Szeged, Szeged, Hungary.,HCEMM-SZTE Skin Research Group, Szeged, Hungary
| | - Edit Urbán
- Department of Public Health, University of Szeged, Szeged, Hungary
| | - Katalin Burián
- Institute of Clinical Microbiology, University of Szeged, Szeged, Hungary
| | - Lajos Kemény
- Department of Dermatology and Allergology, University of Szeged, Szeged, Hungary.,HCEMM-SZTE Skin Research Group, Szeged, Hungary.,MTA-SZTE Dermatological Research Group, Szeged, Hungary
| | - Kornélia Szabó
- Department of Dermatology and Allergology, University of Szeged, Szeged, Hungary. .,MTA-SZTE Dermatological Research Group, Szeged, Hungary.
| |
Collapse
|
35
|
Stolte KN, Pelz C, Yapto CV, Raguse JD, Dommisch H, Danker K. IL-1β strengthens the physical barrier in gingival epithelial cells. Tissue Barriers 2020; 8:1804249. [PMID: 32835592 DOI: 10.1080/21688370.2020.1804249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
Abstract
Periodontitis is one of the most common oral diseases worldwide and is caused by a variety of interactions between oral bacteria and the host. Here, pathogens induce inflammatory host responses that cause the secretion of proinflammatory cytokines such as IL-1β, IL-6, and IL-8 by oral epithelial cells. In various systems, it has been shown that inflammation compromises physical barriers, which enables bacteria to invade the tissue. In this study, we investigated the barrier properties of the oral mucosa under physiological and inflamed conditions. For this purpose, we assessed the influence of IL-1β on the transepithelial electrical resistance and in particular on tight junctions in vitro in human stratified squamous epithelium models. Indirect immunofluorescence and western blot analyses were performed to investigate localization and expression of tight junction proteins in primary gingival cells, immortalized gingival cells and native gingiva. Furthermore, the TEER of gingival keratinocytes was assessed. The results showed that IL-1β led to strengthening of the gingival keratinocyte barrier. This was demonstrated by an increase in TEER, the upregulation of TJ proteins, and an increase in the formation of TJ strands. The IL-1β-mediated upregulation of occludin was prevented by the NF-κB inhibitor BAY 11-7085. These observations provide insights into host responses in the early stages of periodontal disease and offer information about TJ formation in human gingival epithelial cells under physiological and inflammatory conditions. Comprehensive knowledge of the physical barrier during inflammation may help in developing strategies to effectively target the inflammatory barrier to improve the bioavailability of drugs for the treatment of periodontitis.
Collapse
Affiliation(s)
- Kim Natalie Stolte
- Charitá - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Berlin Institute of Health.,Institute for Biochemistry , Berlin, Germany
| | - Carsten Pelz
- Charitá - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Berlin Institute of Health.,Institute for Biochemistry , Berlin, Germany
| | - Cynthia V Yapto
- Charitá - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Berlin Institute of Health.,Institute for Biochemistry , Berlin, Germany
| | - Jan-Dirk Raguse
- Charitá - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Berlin Institute of Health.,Department of Oral and Maxillofacial Surgery, Berlin, Germany
| | - Henrik Dommisch
- Charitá - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Berlin Institute of Health.,Department of Periodontology and Synoptic Dentistry, Berlin, Germany.,Department of Periodontics, University of Washington , Seattle, WA, USA
| | - Kerstin Danker
- Charitá - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Berlin Institute of Health.,Institute for Biochemistry , Berlin, Germany
| |
Collapse
|
36
|
Turner CT, Zeglinski MR, Richardson KC, Santacruz S, Hiroyasu S, Wang C, Zhao H, Shen Y, Sehmi R, Lima H, Gauvreau GM, Granville DJ. Granzyme B Contributes to Barrier Dysfunction in Oxazolone-Induced Skin Inflammation through E-Cadherin and FLG Cleavage. J Invest Dermatol 2020; 141:36-47. [PMID: 32504614 DOI: 10.1016/j.jid.2020.05.095] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 05/13/2020] [Accepted: 05/22/2020] [Indexed: 01/24/2023]
Abstract
Atopic dermatitis (AD) is the most common inflammatory skin condition. Skin barrier dysfunction is of major importance in AD because it facilitates allergen sensitization and systemic allergic responses. Long regarded as a pro-apoptotic protease, emerging studies indicate granzyme B (GzmB) to have extracellular roles involving the proteolytic cleavage of extracellular matrix, cell adhesion proteins, and basement membrane proteins. Minimally expressed in normal skin, GzmB is elevated in AD and is positively correlated with disease severity and pruritus. We hypothesized that GzmB contributes to AD through extracellular protein cleavage. A causative role for GzmB was assessed in an oxazolone-induced murine model of dermatitis, comparing GzmB-/- mice with wild-type mice, showing significant reductions in inflammation, epidermal thickness, and lesion formation in GzmB-/- mice. Topical administration of a small-molecule GzmB inhibitor reduced disease severity compared with vehicle-treated controls. Mechanistically, GzmB impaired epithelial barrier function through E-cadherin and FLG cleavage. GzmB proteolytic activity contributes to impaired epidermal barrier function and represents a valid therapeutic target for AD.
Collapse
Affiliation(s)
- Christopher T Turner
- International Collaboration on Repair Discoveries (ICORD) Centre, Vancouver Coastal Health Research Institute, Vancouver, British Columbia, Canada; Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada; British Columbia Professional Firefighters' Burn and Wound Healing Group, Vancouver, British Columbia, Canada
| | - Matthew R Zeglinski
- International Collaboration on Repair Discoveries (ICORD) Centre, Vancouver Coastal Health Research Institute, Vancouver, British Columbia, Canada; Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada; British Columbia Professional Firefighters' Burn and Wound Healing Group, Vancouver, British Columbia, Canada
| | - Katlyn C Richardson
- International Collaboration on Repair Discoveries (ICORD) Centre, Vancouver Coastal Health Research Institute, Vancouver, British Columbia, Canada; Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada; British Columbia Professional Firefighters' Burn and Wound Healing Group, Vancouver, British Columbia, Canada
| | - Stephanie Santacruz
- International Collaboration on Repair Discoveries (ICORD) Centre, Vancouver Coastal Health Research Institute, Vancouver, British Columbia, Canada; Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada; British Columbia Professional Firefighters' Burn and Wound Healing Group, Vancouver, British Columbia, Canada
| | - Sho Hiroyasu
- International Collaboration on Repair Discoveries (ICORD) Centre, Vancouver Coastal Health Research Institute, Vancouver, British Columbia, Canada; Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada; British Columbia Professional Firefighters' Burn and Wound Healing Group, Vancouver, British Columbia, Canada
| | - Christine Wang
- International Collaboration on Repair Discoveries (ICORD) Centre, Vancouver Coastal Health Research Institute, Vancouver, British Columbia, Canada; Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada; British Columbia Professional Firefighters' Burn and Wound Healing Group, Vancouver, British Columbia, Canada
| | - Hongyan Zhao
- International Collaboration on Repair Discoveries (ICORD) Centre, Vancouver Coastal Health Research Institute, Vancouver, British Columbia, Canada; Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada; British Columbia Professional Firefighters' Burn and Wound Healing Group, Vancouver, British Columbia, Canada
| | - Yue Shen
- International Collaboration on Repair Discoveries (ICORD) Centre, Vancouver Coastal Health Research Institute, Vancouver, British Columbia, Canada; Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada; British Columbia Professional Firefighters' Burn and Wound Healing Group, Vancouver, British Columbia, Canada
| | - Roma Sehmi
- Division of Respirology, Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Hermenio Lima
- Division of Dermatology, Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Gail M Gauvreau
- Division of Respirology, Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - David J Granville
- International Collaboration on Repair Discoveries (ICORD) Centre, Vancouver Coastal Health Research Institute, Vancouver, British Columbia, Canada; Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada; British Columbia Professional Firefighters' Burn and Wound Healing Group, Vancouver, British Columbia, Canada.
| |
Collapse
|
37
|
Campolo M, Casili G, Paterniti I, Filippone A, Lanza M, Ardizzone A, Scuderi SA, Cuzzocrea S, Esposito E. Effect of a Product Containing Xyloglucan and Pea Protein on a Murine Model of Atopic Dermatitis. Int J Mol Sci 2020; 21:ijms21103596. [PMID: 32438777 PMCID: PMC7279434 DOI: 10.3390/ijms21103596] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 04/23/2020] [Accepted: 05/17/2020] [Indexed: 12/12/2022] Open
Abstract
Atopic dermatitis (AD) is a chronic inflammatory disease of the skin, characterized by dryness and more or less severe itching. The etiology of AD is complex and has not been fully clarified, involving genetic susceptibility, immunological abnormalities, epidermal barrier dysfunction, and environmental factors. Xyloglucan (XG) and pea protein (PP) are two compounds of natural origin characterized by the ability to create a physical barrier that protects mucosae membranes, reducing inflammation. The aim of the present study was to evaluate the potential beneficial effects of XG + PP in both a mouse model of AD and Staphylococcus aureus (S. aureus) infection- associated AD. Mice were topically treated with 200 μL of 0.5% oxazolone on the dorsal skin three times a week for AD induction. Mice received XG and PP by topical administration 1 h before oxazolone treatment. In S. aureus infection-associated AD, to induce a superficial superinfection of the skin, mice were also treated with 5 μL of 108 of a culture of S. aureus for 2 weeks; mice superinfected received XG and PP by topical administration 1 h before oxazolone + S. aureus. Four weeks later, the skin was removed for histological and biochemical analysis. Our results demonstrated the protective barrier effects of XG and PP characterized by a reduction in histological tissue changes, mastocyte degranulation, and tight junction permeability in the skin following oxazolone treatment. Moreover, XG + PP was able to preserve filaggrin expression, a hallmark of AD. Our data also support the effectiveness of XG + PP to reduce the damage by superinfection post AD induced by S. aureus. In conclusion, a future product containing XG and PP could be considered as a potentially interesting approach for the treatment of AD.
Collapse
|
38
|
Lipsky ZW, Marques CNH, German GK. Lipid depletion enables permeation of Staphylococcus aureus bacteria through human stratum corneum. Tissue Barriers 2020; 8:1754706. [PMID: 32338129 PMCID: PMC7549745 DOI: 10.1080/21688370.2020.1754706] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Revised: 04/01/2020] [Accepted: 04/06/2020] [Indexed: 02/01/2023] Open
Abstract
Atopic dermatitis (AD) is a chronic inflammatory disease that affects approximately 2-5% of adults worldwide. The pathogenesis of AD continues to be a well-debated point of conjecture, with numerous hypotheses having been proposed. AD conditions are associated with increased populations of Staphylococcus aureus and reduced skin lipids. In this study, we evaluate the ability of S. aureus to permeate across human stratum corneum (SC) exhibiting both normal and depleted lipid conditions consistent with AD. This permeation would enable bacteria to interact with underlying viable epidermal cells, which could serve as a trigger for inflammation and disease onset. Our results indicate that permeation of S. aureus through SC exhibiting normal lipid conditions is not statistically significant. However, bacteria can readily permeate through lipid depleted tissue over a 9-d period. These findings suggest that S. aureus may potentially act as the mechanistic cause, rather than merely the result of AD. ABBREVIATIONS AD: Atopic dermatitis; SC: Stratum Corneum; AMP: Antimicrobial peptide; DIW: Deionized water; PDMS: Polydimethylsiloxane; GFP: Green fluorescent protein; BHI: Brain heart infusion medium.
Collapse
Affiliation(s)
- Zachary W. Lipsky
- Department of Biomedical Engineering, Binghamton University, Binghamton, NY, USA
- Binghamton Biofilm Research Center, Binghamton University, Binghamton, NY, USA
| | - Cláudia N. H. Marques
- Binghamton Biofilm Research Center, Binghamton University, Binghamton, NY, USA
- Department of Biological Sciences, Binghamton University, Binghamton, NY, USA
| | - Guy K. German
- Department of Biomedical Engineering, Binghamton University, Binghamton, NY, USA
- Binghamton Biofilm Research Center, Binghamton University, Binghamton, NY, USA
| |
Collapse
|
39
|
Singh V, Phukan UJ. Interaction of host and Staphylococcus aureus protease-system regulates virulence and pathogenicity. Med Microbiol Immunol 2019; 208:585-607. [PMID: 30483863 DOI: 10.1007/s00430-018-0573-y] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Accepted: 11/22/2018] [Indexed: 02/06/2023]
Abstract
Staphylococcus aureus causes various health care- and community-associated infections as well as certain chronic TH2 driven inflammatory diseases. It is a potent pathogen with serious virulence and associated high morbidity. Severe pathogenicity is accredited to the S. aureus secreted virulence factors such as proteases and host protease modulators. These virulence factors promote adhesion and invasion of bacteria through damage of tight junction barrier and keratinocytes. They inhibit activation and transmigration of various immune cells such as neutrophils (and neutrophil proteases) to evade opsono-phagocytosis and intracellular bacterial killing. Additionally, they protect the bacteria from extracellular killing by disrupting integrity of extracellular matrix. Platelet activation and agglutination is also impaired by these factors. They also block the classical as well as alternative pathways of complement activation and assist in spread of infection through blood and tissue. As these factors are exquisite factors of S. aureus mediated disease development, we have focused on review of diversification of various protease-system associated virulence factors, their structural building, diverse role in disease development and available therapeutic counter measures. This review summarises the role of protease-associated virulence factors during invasion and progression of disease.
Collapse
Affiliation(s)
- Vigyasa Singh
- Molecular Bioprospection Department, CSIR-Central Institute of Medicinal and Aromatic Plants, P.O. CIMAP, Lucknow, 226015, India
| | - Ujjal Jyoti Phukan
- School of Life Science, Jawaharlal Nehru University, New Delhi, 110067, India.
| |
Collapse
|
40
|
McBain AJ, O'Neill CA, Amezquita A, Price LJ, Faust K, Tett A, Segata N, Swann JR, Smith AM, Murphy B, Hoptroff M, James G, Reddy Y, Dasgupta A, Ross T, Chapple IL, Wade WG, Fernandez-Piquer J. Consumer Safety Considerations of Skin and Oral Microbiome Perturbation. Clin Microbiol Rev 2019; 32:e00051-19. [PMID: 31366612 PMCID: PMC6750131 DOI: 10.1128/cmr.00051-19] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Microbiomes associated with human skin and the oral cavity are uniquely exposed to personal care regimes. Changes in the composition and activities of the microbial communities in these environments can be utilized to promote consumer health benefits, for example, by reducing the numbers, composition, or activities of microbes implicated in conditions such as acne, axillary odor, dandruff, and oral diseases. It is, however, important to ensure that innovative approaches for microbiome manipulation do not unsafely disrupt the microbiome or compromise health, and where major changes in the composition or activities of the microbiome may occur, these require evaluation to ensure that critical biological functions are unaffected. This article is based on a 2-day workshop held at SEAC Unilever, Sharnbrook, United Kingdom, involving 31 specialists in microbial risk assessment, skin and oral microbiome research, microbial ecology, bioinformatics, mathematical modeling, and immunology. The first day focused on understanding the potential implications of skin and oral microbiome perturbation, while approaches to characterize those perturbations were discussed during the second day. This article discusses the factors that the panel recommends be considered for personal care products that target the microbiomes of the skin and the oral cavity.
Collapse
Affiliation(s)
- Andrew J McBain
- Division of Pharmacy & Optometry, School of Health Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, United Kingdom
| | - Catherine A O'Neill
- Division of Musculoskeletal & Dermatological Sciences, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, United Kingdom
| | - Alejandro Amezquita
- Unilever, Safety & Environmental Assurance Centre (SEAC), Sharnbrook, United Kingdom
| | - Laura J Price
- Unilever, Safety & Environmental Assurance Centre (SEAC), Sharnbrook, United Kingdom
| | - Karoline Faust
- Department of Microbiology, Immunology and Transplantation, Laboratory of Molecular Bacteriology, Rega Institute, Leuven, Belgium
| | - Adrian Tett
- Department CIBIO, University of Trento, Trento, Italy
| | - Nicola Segata
- Department CIBIO, University of Trento, Trento, Italy
| | - Jonathan R Swann
- Division of Integrative Systems Medicine and Digestive Diseases, Imperial College London, London, United Kingdom
| | | | | | | | | | | | | | - Tom Ross
- University of Tasmania, Hobart, Tasmania, Australia
| | - Iain L Chapple
- Periodontal Research Group, The University of Birmingham, Birmingham, United Kingdom
| | - William G Wade
- Centre for Host-Microbiome Interactions, King's College London, London, United Kingdom
| | | |
Collapse
|
41
|
Renggli K, Rousset N, Lohasz C, Nguyen OTP, Hierlemann A. Integrated Microphysiological Systems: Transferable Organ Models and Recirculating Flow. ADVANCED BIOSYSTEMS 2019; 3:e1900018. [PMID: 32627410 PMCID: PMC7610576 DOI: 10.1002/adbi.201900018] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Revised: 02/28/2019] [Indexed: 01/09/2023]
Abstract
Studying and understanding of tissue and disease mechanisms largely depend on the availability of suitable and representative biological model systems. These model systems should be carefully engineered and faithfully reproduce the biological system of interest to understand physiological effects, pharmacokinetics, and toxicity to better identify new drug compounds. By relying on microfluidics, microphysiological systems (MPSs) enable the precise control of culturing conditions and connections of advanced in vitro 3D organ models that better reproduce in vivo environments. This review focuses on transferable in vitro organ models and integrated MPSs that host these transferable biological units and enable interactions between different tissue types. Interchangeable and transferrable in vitro organ models allow for independent quality control of the biological model before system assembly and building MPS assays on demand. Due to the complexity and different maturation times of individual in vitro tissues, off-chip production and quality control entail improved stability and reproducibility of the systems and results, which is important for large-scale adoption of the technology. Lastly, the technical and biological challenges and open issues for realizing and implementing integrated MPSs with transferable in vitro organ models are discussed.
Collapse
Affiliation(s)
- Kasper Renggli
- ETH Zürich, Department of Biosystems Science and Engineering, Mattenstrasse 26, 4058 Basel, Switzerland
| | | | | | | | | |
Collapse
|
42
|
Faway E, Cambier L, De Vuyst E, Evrard C, Thiry M, Lambert de Rouvroit C, Mignon B, Poumay Y. Responses of Reconstructed Human Epidermis to Trichophyton rubrum Infection and Impairment of Infection by the Inhibitor PD169316. J Invest Dermatol 2019; 139:2080-2089.e6. [PMID: 30986374 DOI: 10.1016/j.jid.2019.03.1147] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Revised: 03/22/2019] [Accepted: 03/25/2019] [Indexed: 12/15/2022]
Abstract
Despite the threatening incidence of dermatophytosis, information is still lacking about the consequences of infection on epidermal barrier functions and about the keratinocyte responses that alert immune components. To identify the mechanisms involved, arthroconidia of the anthropophilic dermatophyte Trichophyton rubrum were prepared to infect reconstructed human epidermis (RHE) in vitro. Integrity of the barrier was monitored during infection by measurements of transepithelial electrical resistance and dye-permeation through the RHE. Expression and release of pro-inflammatory cytokines and antimicrobial peptides by keratinocytes inserted into the RHE were assessed, respectively, by quantitative reverse transcriptase-PCR (to analyze mRNA content in tissue extracts) and by ELISA (to detect proteins in culture media). Results reveal that infection by T. rubrum is responsible for disruption of the epidermal barrier, including loss of functional tight junctions. It additionally causes simultaneous expression and release of cytokines and antimicrobial peptides by keratinocytes. Potential involvement of the p38 mitogen-activated protein kinase signaling pathway was evaluated during infection by targeted inhibition of its activity. Intriguingly, among several p38 mitogen-activated protein kinase inhibitors, PD169316 alone was able to inhibit growth of T. rubrum on Sabouraud agar and to suppress the process of infection on RHE. This suggests that PD169316 acts on a specific target in dermatophytes themselves.
Collapse
Affiliation(s)
- Emilie Faway
- URPHYM-NARILIS, University of Namur, Namur, Belgium
| | - Ludivine Cambier
- FARAH, Faculty of Veterinary Medicine, University of Liège, Liège, Belgium
| | | | | | - Marc Thiry
- Cell and Tissue Biology Unit, GIGA-Neurosciences, University of Liège, Liège, Belgium
| | | | - Bernard Mignon
- FARAH, Faculty of Veterinary Medicine, University of Liège, Liège, Belgium
| | - Yves Poumay
- URPHYM-NARILIS, University of Namur, Namur, Belgium.
| |
Collapse
|
43
|
Shi J, Barakat M, Chen D, Chen L. Bicellular Tight Junctions and Wound Healing. Int J Mol Sci 2018; 19:ijms19123862. [PMID: 30518037 PMCID: PMC6321209 DOI: 10.3390/ijms19123862] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Revised: 11/28/2018] [Accepted: 11/29/2018] [Indexed: 12/15/2022] Open
Abstract
Bicellular tight junctions (TJs) are intercellular junctions comprised of a variety of transmembrane proteins including occludin, claudins, and junctional adhesion molecules (JAMs) as well as intracellular scaffold proteins such as zonula occludens (ZOs). TJs are functional, intercellular structures that form a barrier between adjacent cells, which constantly seals and unseals to control the paracellular passage of molecules. They are primarily present in the epithelial and endothelial cells of all tissues and organs. In addition to their well-recognized roles in maintaining cell polarity and barrier functions, TJs are important regulators of signal transduction, which modulates cell proliferation, migration, and differentiation, as well as some components of the immune response and homeostasis. A vast breadth of research data is available on TJs, but little has been done to decipher their specific roles in wound healing, despite their primary distribution in epithelial and endothelial cells, which are essential contributors to the wound healing process. Some data exists to indicate that a better understanding of the functions and significance of TJs in healing wounds may prove crucial for future improvements in wound healing research and therapy. Specifically, recent studies demonstrate that occludin and claudin-1, which are two TJ component proteins, are present in migrating epithelial cells at the wound edge but are absent in chronic wounds. This indicates that functional TJs may be critical for effective wound healing. A tremendous amount of work is needed to investigate their roles in barrier function, re-epithelialization, angiogenesis, scar formation, and in the interactions between epithelial cells, endothelial cells, and immune cells both in the acute wound healing process and in non-healing wounds. A more thorough understanding of TJs in wound healing may shed new light on potential research targets and reveal novel strategies to enhance tissue regeneration and improve wound repair.
Collapse
Affiliation(s)
- Junhe Shi
- Center for Wound Healing and Tissue Regeneration, College of Dentistry, University of Illinois at Chicago, 801 S. Paulina Street, Chicago, IL 60612, USA.
| | - May Barakat
- Center for Wound Healing and Tissue Regeneration, College of Dentistry, University of Illinois at Chicago, 801 S. Paulina Street, Chicago, IL 60612, USA.
| | - Dandan Chen
- Colgate-Palmolive Company, Piscataway, NJ 08855, USA.
| | - Lin Chen
- Center for Wound Healing and Tissue Regeneration, College of Dentistry, University of Illinois at Chicago, 801 S. Paulina Street, Chicago, IL 60612, USA.
| |
Collapse
|
44
|
Yokouchi M, Kubo A. Maintenance of tight junction barrier integrity in cell turnover and skin diseases. Exp Dermatol 2018; 27:876-883. [DOI: 10.1111/exd.13742] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Revised: 06/29/2018] [Accepted: 07/13/2018] [Indexed: 02/01/2023]
Affiliation(s)
- Mariko Yokouchi
- Department of Dermatology; Keio University School of Medicine; Tokyo Japan
- Nerima General Hospital; Tokyo Japan
| | - Akiharu Kubo
- Department of Dermatology; Keio University School of Medicine; Tokyo Japan
| |
Collapse
|
45
|
Jia Z, Wang X, Wang X, Wei P, Li L, Wu P, Hong M. Calycosin alleviates allergic contact dermatitis by repairing epithelial tight junctions via down-regulating HIF-1α. J Cell Mol Med 2018; 22:4507-4521. [PMID: 29993193 PMCID: PMC6111858 DOI: 10.1111/jcmm.13763] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Accepted: 06/04/2018] [Indexed: 11/28/2022] Open
Abstract
Calycosin, a bioactive component derived from Astragali Radix (AR; Huang Qi), has been shown to have an effect of anti‐allergic dermatitis with unknown mechanism. This study aims to investigate the mechanism of calycosin related to tight junctions (TJs) and HIF‐1α both in FITC‐induced mice allergic contact dermatitis and in IL‐1β stimulated HaCaT keratinocytes. Th2 cytokines (IL‐4, IL‐5 and IL‐13) were detected by ELISA. The epithelial TJ proteins (occludin, CLDN1 and ZO‐1), initiative key cytokines (TSLP and IL‐33) and HIF‐1α were assessed by Western blot, real‐time PCR, immunohistochemistry or immunofluorescence. Herein, we have demonstrated that allergic inflammation and the Th2 cytokines in ACD mice were reduced significantly by calycosin treatment. Meanwhile, calycosin obviously decreased the expression of HIF‐1α and repaired TJs both in vivo and in vitro. In HaCaT keratinocytes, we noted that IL‐1β induced the deterioration of TJs, as well as the increased levels of TSLP and IL‐33, which could be reversed by silencing HIF‐1α. In addition, administration of 2‐methoxyestradiolin (2‐ME), a HIF‐1α inhibitor,significantly repaired the TJs and alleviated the allergic inflammation in vivo. Furthermore, TJs were destroyed by DMOG or by overexpressing HIF‐1α in HaCaT keratinocytes, and simultaneously, calycosin down‐regulated the expression of HIF‐1α and repaired the TJs in this process. These results revealed that calycosin may act as a potential anti‐allergy and barrier‐repair agent via regulating HIF‐1α in AD and suggested that HIF‐1α and TJs might be possible therapy targets for allergic dermatitis.
Collapse
Affiliation(s)
- Zhirong Jia
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Xiaotong Wang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Xiaoyu Wang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Pan Wei
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Lianqu Li
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Peng Wu
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Min Hong
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
46
|
Garcia MA, Nelson WJ, Chavez N. Cell-Cell Junctions Organize Structural and Signaling Networks. Cold Spring Harb Perspect Biol 2018; 10:a029181. [PMID: 28600395 PMCID: PMC5773398 DOI: 10.1101/cshperspect.a029181] [Citation(s) in RCA: 318] [Impact Index Per Article: 45.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Cell-cell junctions link cells to each other in tissues, and regulate tissue homeostasis in critical cell processes that include tissue barrier function, cell proliferation, and migration. Defects in cell-cell junctions give rise to a wide range of tissue abnormalities that disrupt homeostasis and are common in genetic abnormalities and cancers. Here, we discuss the organization and function of cell-cell junctions primarily involved in adhesion (tight junction, adherens junction, and desmosomes) in two different epithelial tissues: a simple epithelium (intestine) and a stratified epithelium (epidermis). Studies in these tissues reveal similarities and differences in the organization and functions of different cell-cell junctions that meet the requirements for the specialized functions of each tissue. We discuss cell-cell junction responses to genetic and environmental perturbations that provide further insights into their roles in maintaining tissue homeostasis.
Collapse
Affiliation(s)
- Miguel A Garcia
- Department of Biology, Stanford University, Stanford, California 94305
| | - W James Nelson
- Department of Biology, Stanford University, Stanford, California 94305
- Departments of Molecular and Cellular Physiology, Stanford University, Stanford, California 94305
| | - Natalie Chavez
- Department of Biology, Stanford University, Stanford, California 94305
| |
Collapse
|
47
|
Cutaneous Barriers and Skin Immunity: Differentiating A Connected Network. Trends Immunol 2018; 39:315-327. [DOI: 10.1016/j.it.2018.02.004] [Citation(s) in RCA: 190] [Impact Index Per Article: 27.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Revised: 02/13/2018] [Accepted: 02/13/2018] [Indexed: 12/12/2022]
|
48
|
Bäsler K, Galliano MF, Bergmann S, Rohde H, Wladykowski E, Vidal-Y-Sy S, Guiraud B, Houdek P, Schüring G, Volksdorf T, Caruana A, Bessou-Touya S, Schneider SW, Duplan H, Brandner JM. Biphasic influence of Staphylococcus aureus on human epidermal tight junctions. Ann N Y Acad Sci 2017; 1405:53-70. [PMID: 28753223 DOI: 10.1111/nyas.13418] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2016] [Revised: 05/23/2017] [Accepted: 05/24/2017] [Indexed: 12/16/2022]
Abstract
Bacterial infections (e.g., with Staphylococcus aureus) are serious problems in skin with a compromised barrier, such as in patients with atopic dermatitis. Previously, it was shown that tight junction (TJ) proteins are influenced by staphylococcal infection, and TJ function is impaired after infection of the keratinocyte cell line HaCaT. However, functional studies in cells or models more similar to human skin are missing. Therefore, we investigated bacterial colonialization and infection with live S. aureus in primary human keratinocytes and reconstructed human epidermis (RHE). We show that short-term inoculation results in increased TJ barrier function-which could not be seen in HaCaT cells-hinting at an early protective effect. This is accompanied by occludin phosphorylation and sustained localization of occludin and claudin-4 at cell membranes. Long-term incubation resulted in decreased presence of claudin-1 and claudin-4 at cell membranes and decreased TJ barrier function. The agr regulon of S. aureus plays a role in the increasing but not in the decreasing effect. Proinflammatory cytokines, which are produced as a result of S. aureus inoculation, influence both phases. In summary, we show here that S. aureus can have short-term promoting effects on the TJ barrier, while in the long term it results in disturbance of TJs.
Collapse
Affiliation(s)
- Katja Bäsler
- Department of Dermatology and Venerology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | | | - Sophia Bergmann
- Department of Dermatology and Venerology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Holger Rohde
- Institute for Medical Microbiology, Virology and Hygiene, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | - Ewa Wladykowski
- Department of Dermatology and Venerology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Sabine Vidal-Y-Sy
- Department of Dermatology and Venerology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | | | - Pia Houdek
- Department of Dermatology and Venerology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Germar Schüring
- Department of Dermatology and Venerology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Thomas Volksdorf
- Department of Dermatology and Venerology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | | | | | - Stefan W Schneider
- Department of Dermatology and Venerology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | | | - Johanna M Brandner
- Department of Dermatology and Venerology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
49
|
Crawford M, Dagnino L. Scaffolding proteins in the development and maintenance of the epidermal permeability barrier. Tissue Barriers 2017; 5:e1341969. [PMID: 28665776 DOI: 10.1080/21688370.2017.1341969] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
The skin of mammals and other terrestrial vertebrates protects the organism against the external environment, preventing heat, water and electrolyte loss, as well as entry of chemicals and pathogens. Impairments in the epidermal permeability barrier function are associated with the genesis and/or progression of a variety of pathological conditions, including genetic inflammatory diseases, microbial and viral infections, and photodamage induced by UV radiation. In mammals, the outside-in epidermal permeability barrier is provided by the joint action of the outermost cornified layer, together with assembled tight junctions in granular keratinocytes found in the layers underneath. Tight junctions serve as both outside-in and inside-out barriers, and impede paracellular movements of ions, water, macromolecules and microorganisms. At the molecular level, tight junctions consist of integral membrane proteins that form an extracellular seal between adjacent cells, and associate with cytoplasmic scaffold proteins that serve as links with the actin cytoskeleton. In this review, we address the roles that scaffold proteins play specifically in the establishment and maintenance of the epidermal permeability barrier, and how various pathologies alter or impair their functions.
Collapse
Affiliation(s)
- Melissa Crawford
- a Department of Physiology and Pharmacology , Children's Health Research Institute and Lawson Health Research Institute, The University of Western Ontario , London , Ontario , Canada
| | - Lina Dagnino
- a Department of Physiology and Pharmacology , Children's Health Research Institute and Lawson Health Research Institute, The University of Western Ontario , London , Ontario , Canada
| |
Collapse
|
50
|
Wang B, McHugh BJ, Qureshi A, Campopiano DJ, Clarke DJ, Fitzgerald JR, Dorin JR, Weller R, Davidson DJ. IL-1β-Induced Protection of Keratinocytes against Staphylococcus aureus-Secreted Proteases Is Mediated by Human β-Defensin 2. J Invest Dermatol 2017; 137:95-105. [PMID: 27702565 PMCID: PMC5176011 DOI: 10.1016/j.jid.2016.08.025] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2016] [Revised: 08/12/2016] [Accepted: 08/23/2016] [Indexed: 12/11/2022]
Abstract
Atopic dermatitis (AD) is a common chronic inflammatory skin disease that results in significant morbidity. A hallmark of AD is disruption of the critical barrier function of upper epidermal layers, causatively linked to environmental stimuli, genetics, and infection, and a critical current target for the development of new therapeutic and prophylactic interventions. Staphylococcus aureus is an AD-associated pathogen producing virulence factors that induce skin barrier disruption in vivo and contribute to AD pathogenesis. We show, using immortalized and primary keratinocytes, that S. aureus protease SspA/V8 is the dominant secreted factor (in laboratory and AD clinical strains of S. aureus) inducing barrier integrity impairment and tight junction damage. V8-induced integrity damage was inhibited by an IL-1β-mediated mechanism, independent of effects on claudin-1. Induction of keratinocyte expression of the antimicrobial/host defense peptide human β-defensin 2 (hBD2) was found to be the mechanism underpinning this protective effect. Endogenous hBD2 expression was required and sufficient for protection against V8 protease-mediated integrity damage, and exogenous application of hBD2 was protective. This modulatory property of hBD2, unrelated to antibacterial effects, gives new significance to the defective induction of hBD2 in the barrier-defective skin lesions of AD and indicates therapeutic potential.
Collapse
Key Words
- ad, atopic dermatitis
- hbd, human β-defensin
- hdp, host defense peptide
- hpek, human primary epidermal keratinocyte
- lps, lipopolysaccharide
- lta, lipoteichoic acid
- oe, overexpressing
- shrna, small hairpin rna
- ssp, staphylococcus aureus serine protease
- tj, tight junction
- vo, vector only
Collapse
Affiliation(s)
- Bingjie Wang
- MRC Centre for Inflammation Research, Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, UK
| | - Brian J McHugh
- MRC Centre for Inflammation Research, Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, UK
| | - Ayub Qureshi
- MRC Centre for Inflammation Research, Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, UK
| | | | - David J Clarke
- School of Chemistry, University of Edinburgh, Edinburgh, UK
| | - J Ross Fitzgerald
- The Roslin Institute and Edinburgh Infectious Diseases, University of Edinburgh, Easter Bush, Midlothian, UK
| | - Julia R Dorin
- MRC Centre for Inflammation Research, Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, UK
| | - Richard Weller
- MRC Centre for Inflammation Research, Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, UK
| | - Donald J Davidson
- MRC Centre for Inflammation Research, Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, UK.
| |
Collapse
|