1
|
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
|
2
|
Lebonvallet N, Catovic C, Feuilloley M, Leschiera R, Reux A, Talagas M, Cousin I, Misery L, Simon E, Chopin S, Gardères J. In vitro and in vivo efficacy of the Active Oligo Skin complex™, a new active ingredient processed from seawater, on multiple parameters of atopic skin. SKIN HEALTH AND DISEASE 2025; 5:22-30. [PMID: 40125013 PMCID: PMC11924379 DOI: 10.1093/skinhd/vzae029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 03/25/2025]
Abstract
Background Different symptoms are associated with atopic skin, including dryness, pruritus and pain, and affect patients' quality of life. The environment, microbiota, epidermis, immune and nerve cells are all implicated in the pathogenesis of atopic skin. Staphylococcus aureus is the focus of particular attention. Epidermis is implicated at multiple levels: inflammatory process, barrier, control of moisture and water loss. Sensory neurons that participate in cutaneous neurogenic inflammation and pruritus are seen as a potential new target. Specific management strategies and new treatments for adults and children are needed to help in more refractory cases. As a baseline of management, guidelines recommend a treatment to moisturize the skin and maintain the skin barrier function, such as an emollient. Objectives To evaluate a new product in vitro and in vivo in order to validate the potential of its use in people with atopic skin or dry skin. Methods A specific mineral composition, Active Oligo Skin complex™, from seawater was developed and included in a balm. The effects of a solution and balm containing the complex were evaluated in vitro on the growth and biofilm formation of Staphylococcus aureus and Staphylococcus epidermidis in different skin models, and in vivo in adult and young volunteers. Results In vitro, the complex modulated bacterial biofilm formation and growth, decreased cytokine [interleukin (IL)-1, IL-6, IL-4] and neuropeptide (substance P) release, and increased the expression of CL1 and CL4. On volunteers with dry skin, the complex had a moisturizing effect after 1 h of application. Dryness and roughness were also reduced in young participants with atopic skin. The balm decreased erythema and pruritus after 21 days of topical application on 60 young participants. On 22 adult participants, stinging score was decreased after -application. Conclusions The Active Oligo Skin complex™ appears to display potent antipruritic and anti-inflammatory activities, both in vitro and in vivo.
Collapse
Affiliation(s)
| | - Chloé Catovic
- Unité de Recherche 4312 Communication Bactérienne et Stratégies Anti-infectieuses (UR CBSA), Université de Rouen Normandie, Evreux, France
| | - Marc Feuilloley
- Unité de Recherche 4312 Communication Bactérienne et Stratégies Anti-infectieuses (UR CBSA), Université de Rouen Normandie, Evreux, France
- Centre d'Expertise Cosmetomics@URN, Université de Rouen Normandie, Evreux, France
| | | | | | | | | | | | - Emilie Simon
- Laboratoires Gilbert, R&D Department, Hérouville St Clair, France
| | - Sylvie Chopin
- Laboratoires Gilbert, R&D Department, Hérouville St Clair, France
| | - Johan Gardères
- Laboratoires Gilbert, R&D Department, Hérouville St Clair, France
| |
Collapse
|
3
|
Burke Ó, Zeden MS, O’Gara JP. The pathogenicity and virulence of the opportunistic pathogen Staphylococcus epidermidis. Virulence 2024; 15:2359483. [PMID: 38868991 PMCID: PMC11178275 DOI: 10.1080/21505594.2024.2359483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Accepted: 05/19/2024] [Indexed: 06/14/2024] Open
Abstract
The pervasive presence of Staphylococcus epidermidis and other coagulase-negative staphylococci on the skin and mucous membranes has long underpinned a casual disregard for the infection risk that these organisms pose to vulnerable patients in healthcare settings. Prior to the recognition of biofilm as an important virulence determinant in S. epidermidis, isolation of this microorganism in diagnostic specimens was often overlooked as clinically insignificant with potential delays in diagnosis and onset of appropriate treatment, contributing to the establishment of chronic infection and increased morbidity or mortality. While impressive progress has been made in our understanding of biofilm mechanisms in this important opportunistic pathogen, research into other virulence determinants has lagged S. aureus. In this review, the broader virulence potential of S. epidermidis including biofilm, toxins, proteases, immune evasion strategies and antibiotic resistance mechanisms is surveyed, together with current and future approaches for improved therapeutic interventions.
Collapse
Affiliation(s)
- Órla Burke
- Microbiology, School of Biological and Chemical Sciences, University of Galway, Galway, Ireland
| | | | - James P. O’Gara
- Microbiology, School of Biological and Chemical Sciences, University of Galway, Galway, Ireland
| |
Collapse
|
4
|
Delanghe L, De Boeck I, Van Malderen J, Gehrmann T, Allonsius CN, Bron PA, Claes I, Hagendorens M, Leysen J, Wittouck S, Lebeer S. The inner elbow skin microbiome contains Lactobacillus among its core taxa and varies with age, season and lifestyle. MICROBIOME RESEARCH REPORTS 2024; 3:43. [PMID: 39741954 PMCID: PMC11684916 DOI: 10.20517/mrr.2024.23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 08/13/2024] [Accepted: 08/22/2024] [Indexed: 01/03/2025]
Abstract
Background: The human skin microbiome plays an essential role in protecting against pathogens and other external substances. This open ecosystem is also influenced by personal and environmental factors, but the precise impact of these factors, such as lifestyle and season, is understudied. We focused here on the inner elbow, a skin site prone to inflammatory conditions like atopic dermatitis and psoriasis. Methods: We collected skin swabs from the inner elbow of 52 children and adults, with no signs of skin disorders, in the winter and summer seasons. Samples were analyzed using metagenomic shallow shotgun sequencing. In addition, metadata were collected using questionnaires on health, lifestyle, and environmental factors. Results: The core inner elbow community, taxa with a prevalence of 95% or higher, consisted of several well-known skin taxa, such as Staphylococcus hominis, Staphylococcus capitis, Staphylococcus epidermidis, and Cutibacterium acnes. In addition, Streptococcus and Lactobacillus species were also found to be highly prevalent members of the skin microbiota, especially in the age group up to 3 years old. Of all investigated factors, age appeared to be the major driver defining the skin microbiome composition and longitudinal stability over the seasons. Differential abundance analysis using three statistical tests also pointed out that specific skin species were significantly associated with sampling season, age, hygiene practices, vitamin D supplements, probiotics, and the number of household members. Conclusion: This study identifies novel factors influencing the inner elbow skin microbiome composition and paves the way for future comparative and intervention studies in skin disorders such as atopic dermatitis.
Collapse
Affiliation(s)
- Lize Delanghe
- Department of Bioscience Engineering, University of Antwerp, Antwerpen 2020, Belgium
| | - Ilke De Boeck
- Department of Bioscience Engineering, University of Antwerp, Antwerpen 2020, Belgium
| | - Joke Van Malderen
- Department of Bioscience Engineering, University of Antwerp, Antwerpen 2020, Belgium
| | - Thies Gehrmann
- Department of Bioscience Engineering, University of Antwerp, Antwerpen 2020, Belgium
| | | | - Peter A. Bron
- Department of Bioscience Engineering, University of Antwerp, Antwerpen 2020, Belgium
| | | | - Margo Hagendorens
- Department of Pediatrics, University Hospital Antwerp/University of Antwerp, Edegem 2650, Belgium
| | - Julie Leysen
- Department of Dermatology, University Hospital Antwerp/University of Antwerp, Edegem 2650, Belgium
| | - Stijn Wittouck
- Department of Bioscience Engineering, University of Antwerp, Antwerpen 2020, Belgium
| | - Sarah Lebeer
- Department of Bioscience Engineering, University of Antwerp, Antwerpen 2020, Belgium
| |
Collapse
|
5
|
Tanishima S, Mihara T, Takeda C, Fujiwara S, Nagashima H. Trends in infectious spondylitis from 2000 to 2020. EUROPEAN SPINE JOURNAL : OFFICIAL PUBLICATION OF THE EUROPEAN SPINE SOCIETY, THE EUROPEAN SPINAL DEFORMITY SOCIETY, AND THE EUROPEAN SECTION OF THE CERVICAL SPINE RESEARCH SOCIETY 2024; 33:3154-3160. [PMID: 38693341 DOI: 10.1007/s00586-024-08286-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Revised: 03/29/2024] [Accepted: 04/18/2024] [Indexed: 05/03/2024]
Abstract
PURPOSE This study aimed to investigate the trends in infectious spondylitis over the past two decades. METHODS We included 157 cases, from 2000 to 2020, of infectious spondylitis. The cases were divided into two groups: 00 (cases during 2000-2009; 82 cases:) and 10 (cases during 2010-2020; 75 cases) groups. Patients' age, sex, causative organism, and localization were examined and compared between the two groups. RESULTS The proportions of women in the 00 and 10 groups were 30.5% and 38.7%, respectively, with no significant difference (P = 0.28). The average age was significantly higher in the 10 group (72.6 years) than in the 00 group (68.8 years; P < 0.01). A compromised host was the cause of infection in 52.4% and 36.0% of the patients in the 00 and 10 groups, respectively, showing a significant difference. The bacterial identification rates were 70.1% and 77.3% in the 00 and 10 groups, respectively (P < 0.01), and the genus Staphylococcus was the most common bacteria. The proportions of resistant bacteria such as methicillin-resistant Staphylococcus aureus in the 00 and 10 groups were 27.3% and 6.7%, respectively (P < 0.01). Conversely, infectious diseases caused by indigenous bacteria in the oral cavity and intestines were more common in the 10group (37.8%) than in the 00 group (13.0%), showing a significant difference (P < 0.01). CONCLUSION Recently, infections caused by indigenous bacteria in the oral cavity and intestines have increased more than those caused by resistant bacteria over the past two decade.
Collapse
Affiliation(s)
- Shinji Tanishima
- Division of Orthopedic Surgery, Department of Sensory and Motor Organs, School of Medicine, Faculty of Medicine, Tottori University, 36-1 Nishi-cho, Yonago, Tottori, 683-8504, Japan.
| | - Tokumitsu Mihara
- Division of Orthopedic Surgery, Department of Sensory and Motor Organs, School of Medicine, Faculty of Medicine, Tottori University, 36-1 Nishi-cho, Yonago, Tottori, 683-8504, Japan
| | - Chikako Takeda
- Division of Orthopedic Surgery, Department of Sensory and Motor Organs, School of Medicine, Faculty of Medicine, Tottori University, 36-1 Nishi-cho, Yonago, Tottori, 683-8504, Japan
| | - Satoshi Fujiwara
- Division of Orthopedic Surgery, Department of Sensory and Motor Organs, School of Medicine, Faculty of Medicine, Tottori University, 36-1 Nishi-cho, Yonago, Tottori, 683-8504, Japan
| | - Hideki Nagashima
- Division of Orthopedic Surgery, Department of Sensory and Motor Organs, School of Medicine, Faculty of Medicine, Tottori University, 36-1 Nishi-cho, Yonago, Tottori, 683-8504, Japan
| |
Collapse
|
6
|
Qu B, Zhang XE, Feng H, Yan B, Bai Y, Liu S, He Y. Microbial perspective on the skin-gut axis and atopic dermatitis. Open Life Sci 2024; 19:20220782. [PMID: 38623584 PMCID: PMC11017189 DOI: 10.1515/biol-2022-0782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 10/26/2023] [Accepted: 10/27/2023] [Indexed: 04/17/2024] Open
Abstract
Atopic dermatitis (AD) is a relapsing inflammatory skin condition that has become a global health issue with complex etiology and mounting prevalence. The association of AD with skin and gut microbiota has been revealed by virtue of the continuous development of sequencing technology and genomics analysis. Also, the gut-brain-skin axis and its mutual crosstalk mechanisms have been gradually verified. Accordingly, the microbiota-skin-gut axis also plays an important role in allergic skin inflammation. Herein, we reviewed the relationship between the microbiota-skin-gut axis and AD, explored the underlying signaling molecules and potential pathways, and focused on the potential mechanisms of probiotics, antimicrobial peptides (AMPs), coagulase-negative staphylococci transplantation, fecal microbiota transplantation, AMPs, and addition of essential fatty acids in alleviating AD, with the aim to provide a new perspective for targeting microbiota in the treatment of allergic skin inflammation.
Collapse
Affiliation(s)
- Bo Qu
- Hospital of Chengdu University of Traditional Chinese Medicine, No. 39 Shi-er-qiao Road, Chengdu, 610072, Sichuan Province, P.R. China
| | - Xue-er Zhang
- Chengdu University of Traditional Chinese Medicine, No. 37 Shi-er-qiao Road, Chengdu, 610072, Sichuan Province, P.R. China
| | - Haoyue Feng
- Hospital of Chengdu University of Traditional Chinese Medicine, No. 39 Shi-er-qiao Road, Chengdu, 610072, Sichuan Province, P.R. China
| | - Bonan Yan
- Chengdu University of Traditional Chinese Medicine, No. 37 Shi-er-qiao Road, Chengdu, 610072, Sichuan Province, P.R. China
| | - Yingchun Bai
- Hospital of Chengdu University of Traditional Chinese Medicine, No. 39 Shi-er-qiao Road, Chengdu, 610072, Sichuan Province, P.R. China
| | - Shanlin Liu
- Chengdu University of Traditional Chinese Medicine, No. 37 Shi-er-qiao Road, Chengdu, 610072, Sichuan Province, P.R. China
| | - Yuhua He
- Hospital of Chengdu University of Traditional Chinese Medicine, No. 39 Shi-er-qiao Road, Chengdu, 610072, Sichuan Province, P.R. China
| |
Collapse
|
7
|
Suellen Ferro de Oliveira C, Kekhasharú Tavaria F. The impact of bioactive textiles on human skin microbiota. Eur J Pharm Biopharm 2023:S0939-6411(23)00118-2. [PMID: 37182552 DOI: 10.1016/j.ejpb.2023.05.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 05/03/2023] [Accepted: 05/05/2023] [Indexed: 05/16/2023]
Abstract
In order to support the elevated market demand for the development of textiles with specific benefits for a healthy and safe lifestyle, several bioactive textiles with defined properties, including antimicrobial, antioxidant, anti-inflammatory, anti-odor, and anti-repellent, anti-ultraviolet (UV) radiation, have been proposed. Antimicrobial textiles, particularly, have received special interest considering the search for smart, protective textiles that also impact health and well-being. Although the incorporation of antimicrobials into textile material has been well succeeded, the addition of such components in textile clothing can influence the balance of the skin microbiota of the wearer. While most antimicrobial textiles have demonstrated good biocompatibility and antimicrobial performance against bacteria, fungi, and viruses, some problems such as textile biodegradation, odor, and dissemination of unwanted microorganisms might arise. However, little is known about the impact of such antimicrobial textile-products on human skin microbiota. To address this issue, the present review, for the first time, gives an overview about the main effects of antimicrobial textiles, i.e., antibacterial, antifungal, and antiviral, on skin microbiota while driving future investigation to elucidate their putative clinical relevance and possible applications according to their impact on skin microbiota. This knowledge may open doors for the development of more microbiota friendly textiles or antimicrobial textile-products able to target specific populations of the skin microbiota aiming to alleviate skin disorders, malodor, and allergies by avoiding the growth and spread of pathogenic microorganisms.
Collapse
Affiliation(s)
- Cláudia Suellen Ferro de Oliveira
- Universidade Católica Portuguesa, CBQF - Centro de Biotecnologia e Química Fina - Laboratório Associado, Escola Superior de Biotecnologia, Rua Diogo Botelho 1327, 4169-005 Porto, Portugal.
| | - Freni Kekhasharú Tavaria
- Universidade Católica Portuguesa, CBQF - Centro de Biotecnologia e Química Fina - Laboratório Associado, Escola Superior de Biotecnologia, Rua Diogo Botelho 1327, 4169-005 Porto, Portugal.
| |
Collapse
|
8
|
Huang C, Li W, Ren X, Tang M, Zhang K, Zhuo F, Dou X, Yu B. The Crucial Roles and Research Advances of cGAS-STING Pathway in Cutaneous Disorders. Inflammation 2023:10.1007/s10753-023-01812-7. [PMID: 37083899 PMCID: PMC10119538 DOI: 10.1007/s10753-023-01812-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 03/14/2023] [Accepted: 03/29/2023] [Indexed: 04/22/2023]
Abstract
The cGAS-STING signaling pathway senses the presence of cytosolic DNA, induces strong type I interferon responses, and enhances inflammatory cytokine production, placing it as an important axis in infection, autoimmunity, and tumor immunity. Recent studies have shown that the abnormalities and/or dysfunctions of cGAS-STING signaling are closely related to the pathogenesis of skin diseases and/or cancers. Additionally, a variety of new therapeutics targeting the cGAS-STING signaling are in development for the treatment of skin disorders. However, the precise molecular mechanisms of cGAS-STING-mediated cutaneous disorders have not been fully elucidated. In this review, we will summarize the regulatory roles and mechanisms of cGAS-STING signaling in skin disorders and recent progresses of cGAS-STING-related drugs as well as their potential clinical applications.
Collapse
Affiliation(s)
- Cong Huang
- Department of Dermatology, Skin Research Institute of Peking University Shenzhen Hospital, Peking University Shenzhen Hospital, Shenzhen Peking University - The Hong Kong University of Science and Technology Medical Center, Shenzhen, 518036, China
| | - Wenting Li
- Department of Dermatology, Skin Research Institute of Peking University Shenzhen Hospital, Peking University Shenzhen Hospital, Shenzhen Peking University - The Hong Kong University of Science and Technology Medical Center, Shenzhen, 518036, China
| | - Xuanyao Ren
- Biomedical Research Institute, Shenzhen Peking University - the Hong Kong University of Science and Technology Medical Center, Shenzhen, 518036, China
| | - Mindan Tang
- Biomedical Research Institute, Shenzhen Peking University - the Hong Kong University of Science and Technology Medical Center, Shenzhen, 518036, China
| | - Kaoyuan Zhang
- Biomedical Research Institute, Shenzhen Peking University - the Hong Kong University of Science and Technology Medical Center, Shenzhen, 518036, China
| | - Fan Zhuo
- Department of Dermatology, Skin Research Institute of Peking University Shenzhen Hospital, Peking University Shenzhen Hospital, Shenzhen Peking University - The Hong Kong University of Science and Technology Medical Center, Shenzhen, 518036, China
| | - Xia Dou
- Department of Dermatology, Skin Research Institute of Peking University Shenzhen Hospital, Peking University Shenzhen Hospital, Shenzhen Peking University - The Hong Kong University of Science and Technology Medical Center, Shenzhen, 518036, China
| | - Bo Yu
- Department of Dermatology, Skin Research Institute of Peking University Shenzhen Hospital, Peking University Shenzhen Hospital, Shenzhen Peking University - The Hong Kong University of Science and Technology Medical Center, Shenzhen, 518036, China.
| |
Collapse
|
9
|
Khelaifia S, Virginie P, Belkacemi S, Tassery H, Terrer E, Aboudharam G. Culturing the Human Oral Microbiota, Updating Methodologies and Cultivation Techniques. Microorganisms 2023; 11:microorganisms11040836. [PMID: 37110259 PMCID: PMC10143722 DOI: 10.3390/microorganisms11040836] [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: 01/24/2023] [Revised: 03/16/2023] [Accepted: 03/23/2023] [Indexed: 04/29/2023] Open
Abstract
Recent years have been marked by a paradigm shift in the study of the human microbiota, with a re-emergence of culture-dependent approaches. Numerous studies have been devoted to the human microbiota, while studies on the oral microbiota still remain limited. Indeed, various techniques described in the literature may enable an exhaustive study of the microbial composition of a complex ecosystem. In this article, we report different methodologies and culture media described in the literature that can be applied to study the oral microbiota by culture. We report on specific methodologies for targeted culture and specific culture techniques and selection methodologies for cultivating members of the three kingdoms of life commonly found in the human oral cavity, namely, eukaryota, bacteria and archaea. This bibliographic review aims to bring together the various techniques described in the literature, enabling a comprehensive study of the oral microbiota in order to demonstrate its involvement in oral health and diseases.
Collapse
Affiliation(s)
- Saber Khelaifia
- Institut Hospitalo-Universitaire Méditerranée-Infection, Aix-Marseille Univ, IRD, MEPHI, AP-HM, 19-21 Boulevard Jean Moulin, 13385 Marseille CEDEX 05, France
- Ecole de Médecine Dentaire, 27 Boulevard Jean Moulin, 13385 Marseille CEDEX 05, France
| | - Pilliol Virginie
- Institut Hospitalo-Universitaire Méditerranée-Infection, Aix-Marseille Univ, IRD, MEPHI, AP-HM, 19-21 Boulevard Jean Moulin, 13385 Marseille CEDEX 05, France
- Ecole de Médecine Dentaire, 27 Boulevard Jean Moulin, 13385 Marseille CEDEX 05, France
| | - Souad Belkacemi
- Institut Hospitalo-Universitaire Méditerranée-Infection, Aix-Marseille Univ, IRD, MEPHI, AP-HM, 19-21 Boulevard Jean Moulin, 13385 Marseille CEDEX 05, France
| | - Herve Tassery
- Ecole de Médecine Dentaire, 27 Boulevard Jean Moulin, 13385 Marseille CEDEX 05, France
| | - Elodie Terrer
- Institut Hospitalo-Universitaire Méditerranée-Infection, Aix-Marseille Univ, IRD, MEPHI, AP-HM, 19-21 Boulevard Jean Moulin, 13385 Marseille CEDEX 05, France
- Ecole de Médecine Dentaire, 27 Boulevard Jean Moulin, 13385 Marseille CEDEX 05, France
| | - Gérard Aboudharam
- Institut Hospitalo-Universitaire Méditerranée-Infection, Aix-Marseille Univ, IRD, MEPHI, AP-HM, 19-21 Boulevard Jean Moulin, 13385 Marseille CEDEX 05, France
- Ecole de Médecine Dentaire, 27 Boulevard Jean Moulin, 13385 Marseille CEDEX 05, France
| |
Collapse
|
10
|
Maynard CL. The Microbiota in Immunity and Inflammation. Clin Immunol 2023. [DOI: 10.1016/b978-0-7020-8165-1.00022-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/08/2023]
|
11
|
Yoshida M, Ishihara T, Isobe Y, Arita M. Genetic deletion of Cyp4f18 disrupts the omega-3 epoxidation pathway and results in psoriasis-like dermatitis. FASEB J 2022; 36:e22648. [PMID: 36374250 DOI: 10.1096/fj.202200982r] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 10/26/2022] [Accepted: 10/31/2022] [Indexed: 11/16/2022]
Abstract
Cyp4f18 catalyzes the conversion of n-3 polyunsaturated fatty acids (PUFAs) into omega-3 epoxides, such as 17,18-epoxyeicosatetraenoic acid (17,18-EpETE) and 19,20-epoxydocosapentaenoic acid (19,20-EpDPE) from eicosapentaenoic acid (EPA), and docosahexaenoic acid (DHA), respectively. Cyp4f18-deficient mice spontaneously develop psoriasis-like dermatitis. A significant increase in the number of IL-17A-positive gamma delta (γδ) T cells in the skin and enlargement of draining lymph nodes was observed. These symptoms were drastically suppressed by antibiotic treatment. Cyp4f18 is highly expressed in dendritic cells (DCs), and Cyp4f18-deficient bone marrow-derived dendritic cells (BMDCs) show markedly increased expression levels of cytokines such as IL-23 and IL-1β in response to lipopolysaccharide (LPS) stimulation. Lipidomic analysis of lymph nodes and BMDCs revealed a significant decrease in a series of omega-3 epoxidized metabolites. Among them, 17,18-dihydroxyeicosatetraenoic acid (17,18-diHETE), a vicinal diol derived from EPA omega-3 epoxidation suppressed IL-23 production in LPS-stimulated BMDCs in Cyp4f18-deficient mice. These results demonstrate that Cyp4f18 endogenously produces omega-3-epoxidized metabolites in the draining lymph nodes, and these metabolites contribute to skin homeostasis by suppressing the excessive activation of the IL-23/IL-17 axis initiated by DCs.
Collapse
Affiliation(s)
- Mio Yoshida
- Division of Physiological Chemistry and Metabolism, Graduate School of Pharmaceutical Sciences, Keio University, Tokyo, Japan.,Laboratory for Metabolomics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Tomoaki Ishihara
- Laboratory for Metabolomics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Yosuke Isobe
- Division of Physiological Chemistry and Metabolism, Graduate School of Pharmaceutical Sciences, Keio University, Tokyo, Japan.,Laboratory for Metabolomics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan.,Cellular and Molecular Epigenetics Laboratory, Graduate School of Medical Life Science, Yokohama City University, Yokohama, Japan
| | - Makoto Arita
- Division of Physiological Chemistry and Metabolism, Graduate School of Pharmaceutical Sciences, Keio University, Tokyo, Japan.,Laboratory for Metabolomics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan.,Cellular and Molecular Epigenetics Laboratory, Graduate School of Medical Life Science, Yokohama City University, Yokohama, Japan
| |
Collapse
|
12
|
Proteomic Analysis of Exudates from Chronic Ulcer of Diabetic Foot Treated with Scorpion Antimicrobial Peptide. Mediators Inflamm 2022; 2022:5852786. [PMID: 36225537 PMCID: PMC9550419 DOI: 10.1155/2022/5852786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 08/26/2022] [Accepted: 09/15/2022] [Indexed: 11/17/2022] Open
Abstract
Scorpion peptides have good therapeutic effect on chronic ulcer of diabetic foot, but the related pharmacological mechanism has remained unclear. The different proteins and bacteria present in ulcer exudates from chronic diabetic foot patients, treated with scorpion antimicrobial peptide at different stages, were analyzed using isobaric tags for quantification-labeled proteomics and bacteriological methods. According to the mass spectrometry data, a total of 1865 proteins were identified qualitatively, and the number of the different proteins was 130 (mid/early), 401 (late/early), and 310 (mid, late/early). In addition, functional annotation, cluster analysis of effects and the analysis of signal pathway, transcription regulation, and protein-protein interaction network were carried out. The results showed that the biochemical changes of wound microenvironment during the treatment involved activated biological functions such as protein synthesis, cell proliferation, differentiation, migration, movement, and survival. Inhibited biological functions such as cell death, inflammatory response, immune diseases, and bacterial growth were also involved. Bacteriological analysis showed that Burkholderia cepacia was the main bacteria in the early and middle stage of ulcer exudate and Staphylococcus epidermidis in the late stage. This study provides basic data for further elucidation of the molecular mechanism of diabetic foot.
Collapse
|
13
|
Characterization of the skin microbiota in bullous pemphigoid patients and controls reveals novel microbial indicators of disease. J Adv Res 2022; 44:71-79. [PMID: 35581140 PMCID: PMC9936408 DOI: 10.1016/j.jare.2022.03.019] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 03/27/2022] [Accepted: 03/31/2022] [Indexed: 02/04/2023] Open
Abstract
INTRODUCTION Bullous pemphigoid (BP) is the most common autoimmune blistering disease. It predominately afflicts the elderly and is significantly associated with increased mortality. The observation of age-dependent changes in the skin microbiota as well as its involvement in other inflammatory skin disorders suggests that skin microbiota may play a role in the emergence of BP blistering. We hypothesize that changes in microbial diversity associated with BP might occur before the emergence of disease lesions, and thus could represent an early indicator of blistering risk. OBJECTIVES The present study aims to investigate potential relationships between skin microbiota and BP and elaborate on important changes in microbial diversity associated with blistering in BP. METHODS The study consisted of an extensive sampling effort of the skin microbiota in patients with BP and age- and sex-matched controls to analyze whether intra-individual, body site, and/or geographical variation correlate with changes in skin microbial composition in BP and/or blistering status. RESULTS We find significant differences in the skin microbiota of patients with BP compared to that of controls, and moreover that disease status rather than skin biogeography (body site) governs skin microbiota composition in patients with BP. Our data reveal a discernible transition between normal skin and the skin surrounding BP lesions, which is characterized by a loss of protective microbiota and an increase in sequences matching Staphylococcus aureus, a known inflammation-promoting species. Notably, Staphylococcus aureus is ubiquitously associated with BP disease status, regardless of the presence of blisters. CONCLUSION The present study suggests Staphylococcus aureus may be a key taxon associated with BP disease status. Importantly, we however find contrasting patterns in the relative abundances of Staphylococcus hominis and Staphylococcus aureus reliably discriminate between patients with BP and matched controls. This may serve as valuable information for assessing blistering risk and treatment outcomes in a clinical setting.
Collapse
|
14
|
Hwang J, Rick J, Hsiao J, Hamzavi IH, Shi VY. Microbiome in Hidradenitis Suppurativa: Current Evidence and Practice. CURRENT DERMATOLOGY REPORTS 2022. [DOI: 10.1007/s13671-021-00349-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
15
|
Determination of drug-resistant bacteria in palmar surface and touchscreen cell phones from bystanders in an urban community. Microbiol Res 2021; 256:126958. [PMID: 34998184 DOI: 10.1016/j.micres.2021.126958] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2021] [Revised: 12/05/2021] [Accepted: 12/27/2021] [Indexed: 11/22/2022]
Abstract
The dynamic microbiota of the human palmar surface (PS) is related to the various hygienic habits of humans and can be transmitted or exchanged upon contact with objects of daily use, such as the indispensable touchscreen cell phone (TCP); this interaction could allow the development of drug-resistant bacteria. The objective was to determine the drug-resistant bacterial contamination between PSs and TCPs in an urban community. Among the total bacterial colonies isolated and molecular and phylogenetically characterized based on the V4-V6 regions of the 16S rRNA gene from PSs and TCPs, the genera present in both types of samples were Staphylococcus (53.3 and 43.5 %, respectively), Bacillus (37.8, 37 %), Atlantibacter (2.2, 10.8 %) and Microbacterium (2.2, 4.3 %). The genera present in only one type of sample were Rothia, Paenibacillus, Escherichia and Micrococcus (2.2 % each). Resistance to penicillins (35.6-93.5 %) and nonsusceptibility to cephalosporins (8.9-37 %) and nitrofurantoin (13.3 and 15.2 %) were observed. The percentage of multidrug antibiotic resistance was 15.4 %. The prevalence of drug-resistant and multidrug-resistant bacteria in PSs and TCPs in the community could give rise to human health problems, and hygiene measures are recommended.
Collapse
|
16
|
Ueta M, Hosomi K, Park J, Mizuguchi K, Sotozono C, Kinoshita S, Kunisawa J. Categorization of the Ocular Microbiome in Japanese Stevens-Johnson Syndrome Patients With Severe Ocular Complications. Front Cell Infect Microbiol 2021; 11:741654. [PMID: 34869055 PMCID: PMC8640524 DOI: 10.3389/fcimb.2021.741654] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Accepted: 10/26/2021] [Indexed: 01/15/2023] Open
Abstract
The commensal microbiota is involved in a variety of diseases. Our group has noticed that patients with Stevens–Johnson syndrome (SJS)/toxic epidermal necrolysis (TEN) often present with persistent inflammation of the ocular surface, even in the chronic stage, and that this inflammation is exacerbated by colonization of the mucosa by certain bacteria. However, the changes in the composition of the ocular microbiome in SJS/TEN patients with severe ocular complications (SOCs) remain to be fully investigated. Here, we conducted a cross-sectional study of 46 Japanese subjects comprising 9 healthy control subjects and 37 SJS/TEN patients with SOC. The 16S rRNA-based genetic analyses revealed that the diversity of the ocular microbiome was reduced in SJS/TEN patients with SOC compared with that in healthy control subjects. Principal coordinate analysis based on Bray–Curtis distance at the genus level revealed that the relative composition of the ocular microbiome was different in healthy control subjects and SJS/TEN patients with SOC, and that the SJS/TEN patients with SOC could be divided into four groups based on whether their microbiome was characterized by enrichment of species in genus Corynebacterium 1, Neisseriaceae uncultured, or Staphylococcus or by simultaneous enrichment in species in genera Propionibacterium, Streptococcus, Fusobacterium, Lawsonella, and Serratia. Collectively, our findings indicate that enrichment of certain bacteria at the ocular surface could be associated with ocular surface inflammation in SJS/TEN patients with SOC.
Collapse
Affiliation(s)
- Mayumi Ueta
- Department of Ophthalmology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Koji Hosomi
- Laboratory of Vaccine Materials, Center for Vaccine and Adjuvant Research and Laboratory of Gut Environmental System, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Ibaraki, Japan
| | - Jonguk Park
- Laboratory of Bioinformatics, Artificial Intelligence Center for Health and Biomedical Research, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Ibaraki, Japan
| | - Kenji Mizuguchi
- Laboratory of Bioinformatics, Artificial Intelligence Center for Health and Biomedical Research, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Ibaraki, Japan.,Institute for Protein Research, Osaka University, Suita, Japan
| | - Chie Sotozono
- Department of Ophthalmology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Shigeru Kinoshita
- Department of Ophthalmology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Jun Kunisawa
- Laboratory of Vaccine Materials, Center for Vaccine and Adjuvant Research and Laboratory of Gut Environmental System, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Ibaraki, Japan.,International Research and Development Center for Mucosal Vaccines, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan.,Graduate School of Medicine, Graduate School of Dentistry, Osaka University, Suita, Japan.,Department of Microbiology and Immunology, Kobe University Graduate School of Medicine, Kobe, Japan
| |
Collapse
|
17
|
Pistone D, Meroni G, Panelli S, D’Auria E, Acunzo M, Pasala AR, Zuccotti GV, Bandi C, Drago L. A Journey on the Skin Microbiome: Pitfalls and Opportunities. Int J Mol Sci 2021; 22:9846. [PMID: 34576010 PMCID: PMC8469928 DOI: 10.3390/ijms22189846] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 09/07/2021] [Accepted: 09/08/2021] [Indexed: 12/22/2022] Open
Abstract
The human skin microbiota is essential for maintaining homeostasis and ensuring barrier functions. Over the years, the characterization of its composition and taxonomic diversity has reached outstanding goals, with more than 10 million bacterial genes collected and cataloged. Nevertheless, the study of the skin microbiota presents specific challenges that need to be addressed in study design. Benchmarking procedures and reproducible and robust analysis workflows for increasing comparability among studies are required. For various reasons and because of specific technical problems, these issues have been investigated in gut microbiota studies, but they have been largely overlooked for skin microbiota. After a short description of the skin microbiota, the review tackles methodological aspects and their pitfalls, covering NGS approaches and high throughput culture-based techniques. Recent insights into the "core" and "transient" types of skin microbiota and how the manipulation of these communities can prevent or combat skin diseases are also covered. Finally, this review includes an overview of the main dermatological diseases, the changes in the microbiota composition associated with them, and the recommended skin sampling procedures. The last section focuses on topical and oral probiotics to improve and maintain skin health, considering their possible applications for skin diseases.
Collapse
Affiliation(s)
- Dario Pistone
- Pediatric Clinical Research Center “Invernizzi”, Department of Biomedical and Clinical Sciences “L. Sacco”, University of Milan, 20157 Milan, Italy; (S.P.); (A.R.P.); (G.V.Z.)
- Department of Biomedical Sciences for Health, University of Milan, 20133 Milan, Italy;
| | - Gabriele Meroni
- Department of Biomedical Surgical and Dental Sciences-One Health Unit, University of Milan, 20133 Milan, Italy;
| | - Simona Panelli
- Pediatric Clinical Research Center “Invernizzi”, Department of Biomedical and Clinical Sciences “L. Sacco”, University of Milan, 20157 Milan, Italy; (S.P.); (A.R.P.); (G.V.Z.)
| | - Enza D’Auria
- Department of Pediatrics, Children’s Hospital Vittore Buzzi, University of Milan, 20154 Milan, Italy; (E.D.); (M.A.)
| | - Miriam Acunzo
- Department of Pediatrics, Children’s Hospital Vittore Buzzi, University of Milan, 20154 Milan, Italy; (E.D.); (M.A.)
| | - Ajay Ratan Pasala
- Pediatric Clinical Research Center “Invernizzi”, Department of Biomedical and Clinical Sciences “L. Sacco”, University of Milan, 20157 Milan, Italy; (S.P.); (A.R.P.); (G.V.Z.)
| | - Gian Vincenzo Zuccotti
- Pediatric Clinical Research Center “Invernizzi”, Department of Biomedical and Clinical Sciences “L. Sacco”, University of Milan, 20157 Milan, Italy; (S.P.); (A.R.P.); (G.V.Z.)
- Department of Pediatrics, Children’s Hospital Vittore Buzzi, University of Milan, 20154 Milan, Italy; (E.D.); (M.A.)
| | - Claudio Bandi
- Pediatric Clinical Research Center “Invernizzi”, Department of Biosciences, University of Milan, 20133 Milan, Italy;
| | - Lorenzo Drago
- Department of Biomedical Sciences for Health, University of Milan, 20133 Milan, Italy;
| |
Collapse
|
18
|
Lee SJ, Kim SE, Shin KO, Park K, Lee SE. Dupilumab Therapy Improves Stratum Corneum Hydration and Skin Dysbiosis in Patients With Atopic Dermatitis. ALLERGY, ASTHMA & IMMUNOLOGY RESEARCH 2021; 13:762-775. [PMID: 34486260 PMCID: PMC8419647 DOI: 10.4168/aair.2021.13.5.762] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 02/15/2021] [Accepted: 03/15/2021] [Indexed: 12/13/2022]
Abstract
Purpose We aimed to investigate the effects of dupilumab on 1) the permeability and antimicrobial barrier, 2) the composition of the skin microbiome, and 3) the correlation between changes in skin barrier properties and microbiota in atopic dermatitis (AD) patients. Methods Ten patients with severe AD were treated with dupilumab for 12 weeks. Disease severity was assessed using the Eczema Area and Severity Index (EASI). Skin barrier function was evaluated by measuring transepidermal water loss, stratum corneum (SC) hydration, and pH. The following parameters were analyzed in the pre- and post-treatment SC samples; 1) skin microbiota using 16S rRNA gene sequencing, 2) lipid composition using mass spectrometry, and 3) human β-defensin 2 (hBD-2) expression using quantitative reverse transcription polymerase chain reaction. Results SC hydration levels in the lesional and non-lesional skin increased after 12-week dupilumab therapy (24.2%, P < 0.001 and 59.9%, P < 0.001, respectively, vs. baseline) and correlated with EASI improvement (r = 0.90, P < 0.001 and r = 0.85, P = 0.003, respectively). Dupilumab increased the long-chain ceramide levels in atopic skin (118.4%, P = 0.028 vs. baseline) that correlated with changes in SC hydration (r = 0.81, P = 0.007) and reduced the elevated hBD-2 messenger RNA levels (−15.4%, P = 0.005 vs. baseline) in the lesional skin. Dupilumab decreased the abundance of Staphylococcus aureus. In contrast, the microbial diversity and the abundance of Cutibacterium and Corynebacterium species increased, which were correlated with an increase in SC hydration levels (Shannon diversity, r = 0.71, P = 0.027; Cutibacterium, r = 0.73, P = 0.017; Corynebacterium, r = 0.75, P = 0.012). Increased abundance of Cutibacterium species was also correlated with EASI improvement (r = 0.68, P = 0.032). Conclusions Th2 blockade-induced normalization of skin microbiome in AD patients is associated with increased SC hydration.
Collapse
Affiliation(s)
- Seung-Ju Lee
- Department of Dermatology and Cutaneous Biology Research Institute, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Song-Ee Kim
- Department of Dermatology and Cutaneous Biology Research Institute, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Kyong-Oh Shin
- Department of Food Science and Nutrition, Convergence Program of Material Science for Medicine and Pharmaceutics, Hallym University, Chuncheon, Korea
| | - Kyungho Park
- Department of Food Science and Nutrition, Convergence Program of Material Science for Medicine and Pharmaceutics, Hallym University, Chuncheon, Korea
| | - Sang Eun Lee
- Department of Dermatology and Cutaneous Biology Research Institute, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea.
| |
Collapse
|
19
|
McLoughlin IJ, Wright EM, Tagg JR, Jain R, Hale JDF. Skin Microbiome-The Next Frontier for Probiotic Intervention. Probiotics Antimicrob Proteins 2021; 14:630-647. [PMID: 34383234 DOI: 10.1007/s12602-021-09824-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/20/2021] [Indexed: 01/16/2023]
Abstract
The skin is the largest organ in the human body, and it orchestrates many functions that are fundamentally important for our survival. Although the skin might appear to present a relatively inhospitable or even hostile environment, a multitude of commensals and also some potentially pathogenic microorganisms have successfully adapted to survive and/or thrive within the diverse ecological niches created by the skin's topographical architecture. Dysbiosis within these microbial populations can result in the emergence and pathological progression of skin diseases. Unsurprisingly, this has led to a new focus of research both for the medical dermatology and cosmetic industries that is concerned with modulation of the skin microbiome to help address common microbially mediated or modulated conditions such as acne, body odour, and atopic dermatitis. This review presents an overview of our current understanding of the complex relationship of the skin with its microbiome and then introduces the concept of probiotic intervention for the management of microbial dysbiosis within the skin ecosystem.
Collapse
Affiliation(s)
| | - Eva M Wright
- School of Pharmacy, University of Otago, PO Box 56, Dunedin, New Zealand
| | - John R Tagg
- Blis Technologies, 81 Glasgow St, South Dunedin, 9012, Dunedin, New Zealand
| | - Rohit Jain
- Blis Technologies, 81 Glasgow St, South Dunedin, 9012, Dunedin, New Zealand
| | - John D F Hale
- Blis Technologies, 81 Glasgow St, South Dunedin, 9012, Dunedin, New Zealand.
| |
Collapse
|
20
|
Nakajima S, Tie D, Nomura T, Kabashima K. Novel pathogenesis of atopic dermatitis from the view of cytokines in mice and humans. Cytokine 2021; 148:155664. [PMID: 34388479 DOI: 10.1016/j.cyto.2021.155664] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 07/21/2021] [Accepted: 07/24/2021] [Indexed: 12/27/2022]
Abstract
Type 2 immunity and inflammation underlie allergic skin disorders, such as atopic dermatitis (AD). In type 2 inflammation, IL-4, IL-13, and IL-5, which are signature type 2 cytokines, are mainly produced by type 2 helper T (Th2) cells and form the characteristic features of AD. Epithelial cell-derived cytokines such as IL-25, IL-33, and TSLP initiate type 2 inflammation by modulating various cells, including group 2 innate lymphoid cells. Moreover, IL-31, a newly identified type 2 cytokine produced mainly by Th2 cells, induces pruritus by acting on sensory neurons in the skin. Based on both basic and clinical findings, several biologics targeting Th2 cytokines have been developed and exhibited significant efficacy as therapeutic reagents for AD. We have summarized the roles of each cytokine (IL-4, 5, 13, 25, 31, and 33, and TSLP) in the development of type 2 inflammation, especially AD, from the view of basic studies in mice and clinical trials/observation in humans.
Collapse
Affiliation(s)
- Saeko Nakajima
- Department of Dermatology, Kyoto University Graduate School of Medicine, Kyoto Japan; Department of Drug Discovery for Inflammatory Skin Diseases, Kyoto University Graduate School of Medicine, Kyoto, Japan.
| | - Duerna Tie
- Department of Dermatology, Kyoto University Graduate School of Medicine, Kyoto Japan
| | - Takashi Nomura
- Department of Dermatology, Kyoto University Graduate School of Medicine, Kyoto Japan
| | - Kenji Kabashima
- Department of Dermatology, Kyoto University Graduate School of Medicine, Kyoto Japan; Singapore Immunology Network (SIgN) and Skin Research Institute of Singapore (SRIS), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore.
| |
Collapse
|
21
|
Sabaté-Brescó M, Berset CM, Zeiter S, Stanic B, Thompson K, Ziegler M, Richards RG, O'Mahony L, Moriarty TF. Fracture biomechanics influence local and systemic immune responses in a murine fracture-related infection model. Biol Open 2021; 10:270855. [PMID: 34240122 PMCID: PMC8496694 DOI: 10.1242/bio.057315] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Accepted: 07/05/2021] [Indexed: 11/26/2022] Open
Abstract
Biomechanical stability plays an important role in fracture healing, with unstable fixation being associated with healing disturbances. A lack of stability is also considered a risk factor for fracture-related infection (FRI), although confirmatory studies and an understanding of the underlying mechanisms are lacking. In the present study, we investigate whether biomechanical (in)stability can lead to altered immune responses in mice under sterile or experimentally inoculated conditions. In non-inoculated C57BL/6 mice, instability resulted in an early increase of inflammatory markers such as granulocyte-colony stimulating factor (G-CSF), keratinocyte chemoattractant (KC) and interleukin (IL)-6 within the bone. When inoculated with Staphylococcus epidermidis, instability resulted in a further significant increase in G-CSF, IL-6 and KC in bone tissue. Staphylococcus aureus infection led to rapid osteolysis and instability in all animals and was not further studied. Gene expression measurements also showed significant upregulation in CCL2 and G-CSF in these mice. IL-17A was found to be upregulated in all S. epidermidis infected mice, with higher systemic IL-17A cell responses in mice that cleared the infection, which was found to be produced by CD4+ and γδ+ T cells in the bone marrow. IL-17A knock-out (KO) mice displayed a trend of delayed clearance of infection (P=0.22, Fisher’s exact test) and an increase in interferon (IFN)-γ production. Biomechanical instability leads to a more pronounced local inflammatory response, which is exaggerated by bacterial infection. This study provides insights into long-held beliefs that biomechanics are crucial not only for fracture healing, but also for control of infection. Summary: Physical movement between bone fragments after a fracture influence healing, and are shown here, for the first time, to influence immune responses and infection.
Collapse
Affiliation(s)
- Marina Sabaté-Brescó
- AO Research Institute Davos, AO Foundation, Davos, Switzerland.,Swiss Institute of Asthma and Allergy Research, University of Zurich, Davos, Switzerland
| | - Corina M Berset
- Swiss Institute of Asthma and Allergy Research, University of Zurich, Davos, Switzerland
| | - Stephan Zeiter
- AO Research Institute Davos, AO Foundation, Davos, Switzerland
| | - Barbara Stanic
- AO Research Institute Davos, AO Foundation, Davos, Switzerland
| | - Keith Thompson
- AO Research Institute Davos, AO Foundation, Davos, Switzerland
| | - Mario Ziegler
- Swiss Institute of Asthma and Allergy Research, University of Zurich, Davos, Switzerland
| | | | - Liam O'Mahony
- Swiss Institute of Asthma and Allergy Research, University of Zurich, Davos, Switzerland
| | | |
Collapse
|
22
|
Interplay between ESKAPE Pathogens and Immunity in Skin Infections: An Overview of the Major Determinants of Virulence and Antibiotic Resistance. Pathogens 2021; 10:pathogens10020148. [PMID: 33540588 PMCID: PMC7912840 DOI: 10.3390/pathogens10020148] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 01/26/2021] [Accepted: 01/27/2021] [Indexed: 12/16/2022] Open
Abstract
The skin is the largest organ in the human body, acting as a physical and immunological barrier against pathogenic microorganisms. The cutaneous lesions constitute a gateway for microbial contamination that can lead to chronic wounds and other invasive infections. Chronic wounds are considered as serious public health problems due the related social, psychological and economic consequences. The group of bacteria known as ESKAPE (Enterococcus faecium, Staphylococcus aureus, Klebsiella pneumoniae, Acinetobacter baumannii, Pseudomonas aeruginosa and Enterobacter sp.) are among the most prevalent bacteria in cutaneous infections. These pathogens have a high level of incidence in hospital environments and several strains present phenotypes of multidrug resistance. In this review, we discuss some important aspects of skin immunology and the involvement of ESKAPE in wound infections. First, we introduce some fundamental aspects of skin physiology and immunology related to cutaneous infections. Following this, the major virulence factors involved in colonization and tissue damage are highlighted, as well as the most frequently detected antimicrobial resistance genes. ESKAPE pathogens express several virulence determinants that overcome the skin's physical and immunological barriers, enabling them to cause severe wound infections. The high ability these bacteria to acquire resistance is alarming, particularly in the hospital settings where immunocompromised individuals are exposed to these pathogens. Knowledge about the virulence and resistance markers of these species is important in order to develop new strategies to detect and treat their associated infections.
Collapse
|
23
|
Haque M, Islam S, Sheikh MA, Dhingra S, Uwambaye P, Labricciosa FM, Iskandar K, Charan J, Abukabda AB, Jahan D. Quorum sensing: a new prospect for the management of antimicrobial-resistant infectious diseases. Expert Rev Anti Infect Ther 2020; 19:571-586. [PMID: 33131352 DOI: 10.1080/14787210.2021.1843427] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Quorum-sensing (QS) is a microbial cell-to-cell communication system that utilizes small signaling molecules to mediates interactions between cross-kingdom microorganisms, including Gram-positive and -negative microbes. QS molecules include N-acyl-homoserine-lactones (AHLs), furanosyl borate, hydroxyl-palmitic acid methylester, and methyl-dodecanoic acid. These signaling molecules maintain the symbiotic relationship between a host and the healthy microbial flora and also control various microbial virulence factors. This manuscript has been developed based on published scientific papers. AREAS COVERED Furanones, glycosylated chemicals, heavy metals, and nanomaterials are considered QS inhibitors (QSIs) and are therefore capable of inhibiting the microbial QS system. QSIs are currently being considered as antimicrobial therapeutic options. Currently, the low speed at which new antimicrobial agents are being developed impairs the treatment of drug-resistant infections. Therefore, QSIs are currently being studied as potential interventions targeting QS-signaling molecules and quorum quenching (QQ) enzymes to reduce microbial virulence. EXPERT OPINION QSIs represent a novel opportunity to combat antimicrobial resistance (AMR). However, no clinical trials have been conducted thus far assessing their efficacy. With the recent advancements in technology and the development of well-designed clinical trials aimed at targeting various components of the, QS system, these agents will undoubtedly provide a useful alternative to treat infectious diseases.
Collapse
Affiliation(s)
- Mainul Haque
- Faculty of Medicine and Defence Health, Universiti Pertahanan Nasional Malaysia (National Defence University of Malaysia), Kuala Lumpur, Malaysia
| | - Salequl Islam
- Department of Microbiology, Jahangirnagar University, Savar, Dhaka, Bangladesh
| | | | - Sameer Dhingra
- School of Pharmacy, Faculty of Medical Sciences, The University of the West Indies, St. Augustine Campus, Eric Williams Medical Sciences Complex, Trinidad & Tobago
| | - Peace Uwambaye
- Department of Preventive & Community Dentistry, University of Rwanda College of Medicine and Health Sciences, School of Dentistry, Kigali, Rwanda
| | | | - Katia Iskandar
- Department of Mathématiques Informatique et Télécommunications, Université Toulouse III, Paul Sabatier, INSERM, UMR 1027, F-31000 Toulouse, France.,INSPECT-LB: Institut National de Santé Publique, d'Épidémiologie Clinique et de Toxicologie-Liban, Beirut 6573-14, Lebanon.,Faculty of Pharmacy, Lebanese University, Beirut 1106, Lebanon
| | - Jaykaran Charan
- Department of Pharmacology, All India Institute of Medical Sciences, Jodhpur, Rajasthan, India
| | | | - Dilshad Jahan
- Department of Hematology, Asgar Ali Hospital, Dhaka, Bangladesh
| |
Collapse
|
24
|
Arteta AA, Milanes-Yearsley M, Cardona-Castro N. Cholangiocyte derived carcinomas and local microbiota. JOURNAL OF HEPATO-BILIARY-PANCREATIC SCIENCES 2020; 29:1084-1093. [PMID: 32902144 DOI: 10.1002/jhbp.826] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 08/10/2020] [Accepted: 08/11/2020] [Indexed: 02/06/2023]
Abstract
Trillions of bacteria are present in the gastrointestinal tract as part of the local microbiota. Bacteria have been associated with a wide range of gastrointestinal diseases including malignant neoplasms. The association of bacteria in gastrointestinal and biliary tract carcinogenesis is supported in the paradigm of Helicobacter pylori and intestinal-type gastric cancer. However, the association of bacterial species to a specific carcinoma, different from intestinal-type gastric cancer is unresolved. The relationship of bacteria to a specific malignant neoplasm can drive clinical interventions. We review the classic bacteria risk factors identified using cultures and PCR (polymerase chain reaction) with new research regarding a microbiota approach through 16S rRNA (16S ribosomal ribonucleic acid gene) or metagenomic analysis for selected carcinomas in the biliary tract.
Collapse
Affiliation(s)
- Ariel A Arteta
- Department of Pathology, University of Antioquia, Medellín, Colombia.,Basic Science Research Group, School of Medicine, CES University, Medellín (Antioquia), Colombia.,Grupo de Investigaciones en Patología, Universidad de Antioquia (GRIP-UdeA), Medellín (Antioquia), Colombia
| | - Martha Milanes-Yearsley
- Ohio State University, Columbus, OH, USA.,Gastrointestinal and Liver Pathology Department, Wexner Medical Center, Columbus, OH, USA
| | | |
Collapse
|
25
|
Riverain-Gillet É, Guet-Revillet H, Jais JP, Ungeheuer MN, Duchatelet S, Delage M, Lam T, Hovnanian A, Nassif A, Join-Lambert O. The Surface Microbiome of Clinically Unaffected Skinfolds in Hidradenitis Suppurativa: A Cross-Sectional Culture-Based and 16S rRNA Gene Amplicon Sequencing Study in 60 Patients. J Invest Dermatol 2020; 140:1847-1855.e6. [DOI: 10.1016/j.jid.2020.02.046] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2019] [Revised: 02/05/2020] [Accepted: 02/16/2020] [Indexed: 12/20/2022]
|
26
|
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
|
27
|
Assessing similarities and disparities in the skin microbiota between wild and laboratory populations of house mice. ISME JOURNAL 2020; 14:2367-2380. [PMID: 32518248 PMCID: PMC7490391 DOI: 10.1038/s41396-020-0690-7] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Revised: 05/15/2020] [Accepted: 05/22/2020] [Indexed: 12/23/2022]
Abstract
The house mouse is a key model organism in skin research including host–microbiota interactions, yet little is known about the skin microbiota of free-living mice. It is similarly unclear how closely laboratory mice, which typically live under exceptionally hygienic conditions, resemble the ancestral state of microbial variation in the wild. In this study, we sampled an area spanning 270 km2 in south-west France and collected 203 wild Mus musculus domesticus. We profiled the ear skin microbiota on standing and active communities (DNA-based and RNA-based 16 rRNA gene sequencing, respectively), and compared multiple community aspects between wild-caught and laboratory-reared mice kept in distinct facilities. Compared to lab mice, we reveal the skin microbiota of wild mice on the one hand to be unique in their composition within the Staphylococcus genus, with a majority of sequences most closely matching known novobiocin-resistant species, and display evidence of a rare biosphere. On the other hand, despite drastic disparities between natural and laboratory environments, we find that shared taxa nonetheless make up the majority of the core skin microbiota of both wild- and laboratory skin communities, suggesting that mammalian skin is a highly specialized habitat capable of strong selection from available species pools. Finally, the influence of environmental factors suggests RNA-based profiling as a preferred method to reduce environmental noise.
Collapse
|
28
|
Nakajima S, Kabata H, Kabashima K, Asano K. Anti-TSLP antibodies: Targeting a master regulator of type 2 immune responses. Allergol Int 2020; 69:197-203. [PMID: 31974038 DOI: 10.1016/j.alit.2020.01.001] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 11/08/2019] [Accepted: 12/10/2019] [Indexed: 12/21/2022] Open
Abstract
TSLP is an epithelial cell-derived cytokine synthesized in response to various stimuli, including protease allergens and microorganisms like viruses and bacteria. Biological functions of TSLP require heterodimer formation between the TSLP receptor (TSLPR) and IL-7 receptor-α, which polarize dendritic cells to induce type 2 inflammation and directly expand and/or activate Th2 cells, group 2 innate lymphoid cells, basophils, and other immune cells. TSLP is thus considered a master regulator of type 2 immune responses at the barrier surfaces of skin and the respiratory/gastrointestinal tract. Indeed, genetic, experimental, and clinical evidence suggests that the TSLP-TSLPR pathway is associated with the pathogenesis of allergic diseases such as atopic dermatitis (AD) and asthma. Tezepelumab (AMG-157/MEDI9929) is a human anti-TSLP antibody that prevents TSLP-TSLPR interactions. A phase 2 trial for moderate to severe AD showed that a greater but not statistically significant percentage of tezepelumab-treated patients showed clinical improvements compared to the placebo group. A phase 2 trial for uncontrolled, severe asthma showed significant decreases in asthma exacerbation rate and improved pulmonary function and asthma control for tezepelumab-treated patients. Levels of biomarkers of type 2 inflammation, such as blood/sputum eosinophil counts and fraction of exhaled nitric oxide decreased, however, clinical efficacy was observed irrespective of the baseline levels of these biomarkers. A blockade of the TSLP-TSLPR pathway likely will exert significant clinical effects on AD, asthma, and other allergic diseases. The efficacy of anti-TSLP antibodies compared to other biologics needs to be further examined.
Collapse
|
29
|
|
30
|
Szari S, Quinn JA. Supporting a Healthy Microbiome for the Primary Prevention of Eczema. Clin Rev Allergy Immunol 2020; 57:286-293. [PMID: 31309394 DOI: 10.1007/s12016-019-08758-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Eczema is increasing worldwide with associated increases in health costs and decreases in quality of life. There are many factors that are speculated to interact in the development of eczema including genetics and environmental exposures. Prevention of the development of eczema may prevent the further development of food allergies and asthma. This concept has prompted a variety of research into the area of primary prevention of eczema in infants. This exploration includes a growing body of research examining infants supplemented with probiotics, prebiotics, or both (synbiotics) often compared with their breastfed counterparts. The goal of this paper is to examine the evidence for manipulating the microbiome in the prevention of eczema. Several strains of probiotics, compositions of prebiotics, and varied combinations of both are commercially available. Evidence supports altering the microbiome in infants at high risk of atopy who are not able to breastfeed with Lactobacillus strains when given both prenatally followed by prolonged use (greater than 6 months) postnatally for the primary prevention of eczema. Prebiotics have also been shown beneficial for primary prevention of eczema in formula-fed infants with prolonged use greater than 6 months. These findings are in keeping with the World Allergy Organization (WAO) recommendations that support interventions to manipulate the microbiome with both probiotics and prebiotics.
Collapse
Affiliation(s)
- Sofia Szari
- Department of Allergy-Immunology, Wilford Hall Ambulatory Surgical Center, Lackland Air Force Base, San Antonio, TX, USA.
| | - James A Quinn
- Department of Allergy-Immunology, Wilford Hall Ambulatory Surgical Center, Lackland Air Force Base, San Antonio, TX, USA
| |
Collapse
|
31
|
Nakajima S, Nomura T, Common J, Kabashima K. Insights into atopic dermatitis gained from genetically defined mouse models. J Allergy Clin Immunol 2019; 143:13-25. [PMID: 30612664 DOI: 10.1016/j.jaci.2018.11.014] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Revised: 11/14/2018] [Accepted: 11/16/2018] [Indexed: 01/01/2023]
Abstract
Atopic dermatitis (AD) is characterized by severe pruritus and recurrent eczema with a chronic disease course. Impaired skin barrier function, hyperactivated TH2 cell-type inflammation, and pruritus-induced scratching contribute to the disease pathogenesis of AD. Skin microbial alterations complicate the pathogenesis of AD further. Mouse models are a powerful tool to analyze such intricate pathophysiology of AD, with a caution that anatomy and immunology of the skin differ between human subjects and mice. Here we review recent understanding of AD etiology obtained using mouse models, which address the epidermal barrier, skin microbiome, TH2 immune response, and pruritus.
Collapse
Affiliation(s)
- Saeko Nakajima
- Department of Dermatology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Takashi Nomura
- Department of Dermatology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - John Common
- Skin Research Institute of Singapore (SRIS), Singapore.
| | - Kenji Kabashima
- Department of Dermatology, Kyoto University Graduate School of Medicine, Kyoto, Japan; Skin Research Institute of Singapore (SRIS), Singapore; Singapore Immunology Network, A*STAR, Singapore.
| |
Collapse
|
32
|
Claudel JP, Auffret N, Leccia MT, Poli F, Corvec S, Dréno B. Staphylococcus epidermidis: A Potential New Player in the Physiopathology of Acne? Dermatology 2019; 235:287-294. [DOI: 10.1159/000499858] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Accepted: 03/25/2019] [Indexed: 11/19/2022] Open
Abstract
Background: Cutibacterium acnes has been identified as one of the main triggers of acne. However, increasing knowledge of the human skin microbiome raises questions about the role of other skin commensals, such as Staphylococcus epidermidis, in the physiopathology of this skin disease. Summary: This review provides an overview of current knowledge of the potential role of S. epidermidis in the physiopathology of acne. Recent research indicates that acne might be the result of an unbalanced equilibrium between C. acnes and S. epidermidis,according to dedicated interactions. Current treatments act on C. acnesonly. Other treatment options may be considered, such as probiotics derived from S. epidermidis to restore the naturally balanced microbiota or through targeting the regulation of the host’s AMP mediators. Key Messages: Research seems to confirm the beneficial role of S. epidermidis in acne by limiting C. acnes over-colonisation and inflammation.
Collapse
|
33
|
Borbón TY, Scorza BM, Clay GM, Lima Nobre de Queiroz F, Sariol AJ, Bowen JL, Chen Y, Zhanbolat B, Parlet CP, Valadares DG, Cassel SL, Nauseef WM, Horswill AR, Sutterwala FS, Wilson ME. Coinfection with Leishmania major and Staphylococcus aureus enhances the pathologic responses to both microbes through a pathway involving IL-17A. PLoS Negl Trop Dis 2019; 13:e0007247. [PMID: 31107882 PMCID: PMC6527190 DOI: 10.1371/journal.pntd.0007247] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Accepted: 02/15/2019] [Indexed: 12/12/2022] Open
Abstract
Cutaneous leishmaniasis (CL) is a parasitic disease causing chronic, ulcerating skin lesions. Most humans infected with the causative Leishmania protozoa are asymptomatic. Leishmania spp. are usually introduced by sand flies into the dermis of mammalian hosts in the presence of bacteria from either the host skin, sand fly gut or both. We hypothesized that bacteria at the dermal inoculation site of Leishmania major will influence the severity of infection that ensues. A C57BL/6 mouse ear model of single or coinfection with Leishmania major, Staphylococcus aureus, or both showed that single pathogen infections caused localized lesions that peaked after 2–3 days for S. aureus and 3 weeks for L. major infection, but that coinfection produced lesions that were two-fold larger than single infection throughout 4 weeks after coinfection. Coinfection increased S. aureus burdens over 7 days, whereas L. major burdens (3, 7, 28 days) were the same in singly and coinfected ears. Inflammatory lesions throughout the first 4 weeks of coinfection had more neutrophils than did singly infected lesions, and the recruited neutrophils from early (day 1) lesions had similar phagocytic and NADPH oxidase capacities. However, most neutrophils were apoptotic, and transcription of immunomodulatory genes that promote efferocytosis was not upregulated, suggesting that the increased numbers of neutrophils may, in part, reflect defective clearance and resolution of the inflammatory response. In addition, the presence of more IL-17A-producing γδ and non-γδ T cells in early lesions (1–7 days), and L. major antigen-responsive Th17 cells after 28 days of coinfection, with a corresponding increase in IL-1β, may recruit more naïve neutrophils into the inflammatory site. Neutralization studies suggest that IL-17A contributed to an enhanced inflammatory response, whereas IL-1β has an important role in controlling bacterial replication. Taken together, these data suggest that coinfection of L. major infection with S. aureus exacerbates disease, both by promoting more inflammation and neutrophil recruitment and by increasing neutrophil apoptosis and delaying resolution of the inflammatory response. These data illustrate the profound impact that coinfecting microorganisms can exert on inflammatory lesion pathology and host adaptive immune responses. Cutaneous leishmaniasis (CL) is a vector-borne ulcerating skin disease affecting several million people worldwide. The causative Leishmania spp. protozoa are transmitted by infected phlebotomine sand flies. During a sand fly bite, bacteria can be coincidentally inoculated into the dermis with the parasite. Staphylococcus aureus is the most common bacterium in CL skin lesions. Symptomatic CL is characterized by papulonodular skin lesions that ulcerate and resolve with scarring, although most cutaneous Leishmania infections are asymptomatic. We sought to explore factors that determine whether infection with a cutaneous Leishmania species would result in symptomatic CL rather than asymptomatic infection. We hypothesized that local bacteria promote the development of symptomatic CL lesions during infection with Leishmania major. We discovered that cutaneous lesions were significantly larger in mice inoculated simultaneously with S. aureus and L. major than in mice infected with either organism alone. Coinfection led to increased S. aureus growth in skin lesions, whereas L. major parasite numbers were unchanged by coinfection. The size of the exacerbated lesion correlated with early increased numbers of neutrophils and elevated levels of proinflammatory cytokines IL-1β and IL-17A during the first 7 days, and with sustained increases in IL-17A through 28 days of coinfection. Neutralizing antibody experiments suggested IL-17A was partially responsible for lesion exacerbation during coinfection, whereas IL-1β was important for both control of early lesion exacerbation and promotion of IL-17A production. These data suggest that treatment of symptomatic CL targeting the parasite, local commensal bacteria, and host proinflammatory IL-17A immune responses might improve the outcome of CL.
Collapse
Affiliation(s)
- Tiffany Y. Borbón
- Department of Microbiology and Immunology, University of Iowa, Iowa City, IA, United States of America
- Medical Scientist Training Program and the Carver College of Medicine, University of Iowa, Iowa City, IA, United States of America
| | - Breanna M. Scorza
- Interdisciplinary Ph.D. Program in Immunology, University of Iowa, Iowa City, IA, United States of America
| | - Gwendolyn M. Clay
- Medical Scientist Training Program and the Carver College of Medicine, University of Iowa, Iowa City, IA, United States of America
- Interdisciplinary Ph.D. Program in Molecular Medicine, University of Iowa, Iowa City, IA, United States of America
| | | | - Alan J. Sariol
- Interdisciplinary Ph.D. Program in Immunology, University of Iowa, Iowa City, IA, United States of America
| | - Jayden L. Bowen
- Medical Scientist Training Program and the Carver College of Medicine, University of Iowa, Iowa City, IA, United States of America
| | - Yani Chen
- Department of Internal Medicine, University of Iowa, Iowa City, Iowa City, IA, United States of America
- Iowa Inflammation Program, Department of Internal Medicine, University of Iowa, Iowa City, IA, United States of America
- Veterans’ Affairs Medical Center, Iowa City, IA, United States of America
| | - Bayan Zhanbolat
- Department of Internal Medicine, University of Iowa, Iowa City, Iowa City, IA, United States of America
- Iowa Inflammation Program, Department of Internal Medicine, University of Iowa, Iowa City, IA, United States of America
- Veterans’ Affairs Medical Center, Iowa City, IA, United States of America
| | - Corey P. Parlet
- Department of Microbiology and Immunology, University of Iowa, Iowa City, IA, United States of America
- Department of Internal Medicine, University of Iowa, Iowa City, Iowa City, IA, United States of America
- Iowa Inflammation Program, Department of Internal Medicine, University of Iowa, Iowa City, IA, United States of America
| | - Diogo G. Valadares
- Department of Microbiology and Immunology, University of Iowa, Iowa City, IA, United States of America
- Department of Internal Medicine, University of Iowa, Iowa City, Iowa City, IA, United States of America
- Iowa Inflammation Program, Department of Internal Medicine, University of Iowa, Iowa City, IA, United States of America
- Veterans’ Affairs Medical Center, Iowa City, IA, United States of America
- Conselho Nacional de Desenvolvimento Cientifico e Tecnológico (CNPq), Brasilia, Brazil
| | - Suzanne L. Cassel
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, United States of America
| | - William M. Nauseef
- Department of Microbiology and Immunology, University of Iowa, Iowa City, IA, United States of America
- Department of Internal Medicine, University of Iowa, Iowa City, Iowa City, IA, United States of America
- Iowa Inflammation Program, Department of Internal Medicine, University of Iowa, Iowa City, IA, United States of America
- Veterans’ Affairs Medical Center, Iowa City, IA, United States of America
| | - Alexander R. Horswill
- Department of Immunology and Microbiology, University of Colorado Denver—Anschutz Medical Campus, Aurora, CO, United States of America
| | - Fayyaz S. Sutterwala
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, United States of America
| | - Mary E. Wilson
- Department of Microbiology and Immunology, University of Iowa, Iowa City, IA, United States of America
- Medical Scientist Training Program and the Carver College of Medicine, University of Iowa, Iowa City, IA, United States of America
- Interdisciplinary Ph.D. Program in Immunology, University of Iowa, Iowa City, IA, United States of America
- Interdisciplinary Ph.D. Program in Molecular Medicine, University of Iowa, Iowa City, IA, United States of America
- Department of Internal Medicine, University of Iowa, Iowa City, Iowa City, IA, United States of America
- Iowa Inflammation Program, Department of Internal Medicine, University of Iowa, Iowa City, IA, United States of America
- Veterans’ Affairs Medical Center, Iowa City, IA, United States of America
- * E-mail:
| |
Collapse
|
34
|
Diaz A, Guttman-Yassky E. Topical agents for the treatment of atopic dermatitis. Expert Rev Clin Immunol 2019; 15:369-382. [PMID: 30587053 DOI: 10.1080/1744666x.2019.1564038] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Accepted: 12/24/2018] [Indexed: 01/11/2023]
Abstract
INTRODUCTION Atopic dermatitis (AD) is perhaps the most common inflammatory skin disorder worldwide, with an increasing incidence in developed countries. The mainstay treatment for patients with AD is topical therapies, which are used not only by the mild patients but also by the moderate-to-severe patients, in conjunction with systemic treatment. While topical steroids and calcineurin antagonists are widely used, these are associated with long-term cutaneous adverse effects (AEs) or a black box warning, preventing their chronic use. Areas covered: The aim of this review is to provide a comprehensive overview of new and upcoming topical therapies currently in development and undergoing clinical trials, as well as their safety and efficacy profiles, and discuss current topicals used in the management of AD. Expert opinion: AD is a heterogeneous disease with complex pathophysiology. Treatments available to date for AD provide disease control; however, patients struggle to find an optimized therapeutic regimen they may use long term and without severe effects. Novel therapies are currently under investigation, with the hope of shifting the paradigm of AD management from symptom control to disease eradication.
Collapse
Affiliation(s)
- Aisleen Diaz
- a Department of Dermatology , Icahn School of Medicine , New York , NY , USA
- b Laboratory for Inflammatory Skin Diseases , New York , NY , USA
- c Ponce Health Sciences University School of Medicine , Ponce , PR , USA
| | - Emma Guttman-Yassky
- a Department of Dermatology , Icahn School of Medicine , New York , NY , USA
- b Laboratory for Inflammatory Skin Diseases , New York , NY , USA
| |
Collapse
|
35
|
|
36
|
Meunier M, Scandolera A, Chapuis E, Lambert C, Jarrin C, Robe P, Chajra H, Auriol D, Reynaud R. From stem cells protection to skin microbiota balance: Orobanche rapum extract, a new natural strategy. J Cosmet Dermatol 2018; 18:1140-1154. [PMID: 30485658 PMCID: PMC7379634 DOI: 10.1111/jocd.12804] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Accepted: 09/17/2018] [Indexed: 12/25/2022]
Abstract
Background Healthy skin is a delicate balance between skin renewal and microbiota homeostasis, and its imbalance promotes premature aging and dermatological disorders. Skin stem cells are key actors in this process but their sensitivity to aging and external stressors such as UV reduces the skin renewal power. The skin microbiota has been recently described as active in the healthy skin, and its imbalance could trigger some disorders. Aims We hypothesized that reactivation of stem cells and maintenance of microbiota could be a disruptive strategy for younger and healthier skin. We thus developed a new plant extract that restores the entire skin renewal process by sequential activation from stem cells stimulation to microbiota protection. Methods We studied stem cells comportment in the presence of Orobanche rapum extract by survivin immunocytochemistry and caspases 3 and 9 dosages. We also analyzed epidermal differentiation markers by immunohistochemistry and lipids organization by GC/MS At the clinical level, we investigated the impact of O. rapum extract on microbiota and on skin aspect. Results We demonstrated an active protection of skin stem cells through the maintenance of their clone‐forming capacity and resistance to UV through the overexpression of survivin coupled to caspases inhibition. Furthermore, we showed the restoration of epidermal differentiation markers and ceramide biosynthesis favorable to orthorhombic organization. Clinical studies, including microbiota analysis, showed an active skin surface renewal coupled with microbiota protection. Conclusion We evidenced that our active ingredient is able to stimulate skin rejuvenation while protecting the cutaneous microbiota, creating healthier skin and thereby beauty.
Collapse
Affiliation(s)
- Marie Meunier
- Research and Development, Givaudan France SAS, Pomacle, France
| | | | - Emilie Chapuis
- Research and Development, Givaudan France SAS, Pomacle, France
| | - Carole Lambert
- Research and Development, Givaudan France SAS, Toulouse, France
| | - Cyrille Jarrin
- Research and Development, Givaudan France SAS, Toulouse, France
| | - Patrick Robe
- Research and Development, Givaudan France SAS, Toulouse, France
| | - Hanane Chajra
- Research and Development, Givaudan France SAS, Toulouse, France
| | - Daniel Auriol
- Research and Development, Givaudan France SAS, Toulouse, France
| | - Romain Reynaud
- Research and Development, Givaudan France SAS, Pomacle, France
| |
Collapse
|
37
|
Strzępa A, Lobo FM, Majewska-Szczepanik M, Szczepanik M. Antibiotics and autoimmune and allergy diseases: Causative factor or treatment? Int Immunopharmacol 2018; 65:328-341. [PMID: 30359934 DOI: 10.1016/j.intimp.2018.10.021] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Revised: 10/11/2018] [Accepted: 10/14/2018] [Indexed: 02/06/2023]
Abstract
The newborn infant emerges from an almost sterile environment into a world of bacteria. Bacteria colonize the infant's skin, lungs, and, of most importance, the gut. The process of bacterial colonization is coordinated, and each body niche acquires a unique composition of bacteria. In the gut, most bacteria belong to the Firmicutes and Bacteroidetes phyla, while Actinobacteria and Proteobacteria are far less abundant. Some of these bacteria possess strong immunoregulatory properties. Bacterial colonization is essential to skew the newborn's immune response away from the allergy-favoring Type-2 response towards a Type-1 immune response, which is essential for pathogen elimination. Imbalance between Type 1 and Type 2 responses, however, can promote autoimmunity. In addition, the microbiota shapes immune responses in adults. Autoimmune and allergic diseases are commonly associated with an altered composition of resident bacteria, which is known as dysbiosis. Perhaps the most common cause of disruption and alteration of the bacterial colonization of newborns is the use of antibiotics. It is not known whether the dysbiosis precedes or is the consequence of allergic and autoimmune disorders, and whether antibiotics can be a trigger for these disorders, depending on the type of antibiotic used and the maturity of immune system. In this review, we discuss the development of the microbiota in different body niches and their immunomodulatory potential. We evaluate the impact of antibiotics, both in mice and in humans, on microbial communities and how that may impact the development and manifestation of diseases through all stages of life: the prenatal period, childhood, and adulthood.
Collapse
Affiliation(s)
- Anna Strzępa
- Department of Medical Biology, Faculty of Health Sciences, Jagiellonian University Medical College, ul. Kopernika 7a, 31-034 Krakow, Poland
| | - Francis M Lobo
- Section of Allergy and Clinical Immunology, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Monika Majewska-Szczepanik
- Department of Medical Biology, Faculty of Health Sciences, Jagiellonian University Medical College, ul. Kopernika 7a, 31-034 Krakow, Poland
| | - Marian Szczepanik
- Department of Medical Biology, Faculty of Health Sciences, Jagiellonian University Medical College, ul. Kopernika 7a, 31-034 Krakow, Poland.
| |
Collapse
|
38
|
Adaptive Metabolism in Staphylococci: Survival and Persistence in Environmental and Clinical Settings. J Pathog 2018; 2018:1092632. [PMID: 30327733 PMCID: PMC6171259 DOI: 10.1155/2018/1092632] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Revised: 07/28/2018] [Accepted: 08/12/2018] [Indexed: 01/04/2023] Open
Abstract
Staphylococci are highly successful at colonizing a variety of dynamic environments, both nonpathogenic and those of clinical importance, and comprise the list of pathogens of global public health significance. Their remarkable survival and persistence can be attributed to a host of strategies, one of which is metabolic versatility—their ability to rapidly alter their metabolism in the presence of transient or long-term bacteriostatic and bactericidal conditions and facilitate cellular homeostasis. These attributes contribute to their widespread dissemination and challenging eradication particularly from clinical settings. The study of microbial behaviour at the metabolite level provides insight into mechanisms of survival and persistence under defined environmental and clinical conditions. This paper reviews the range of metabolic modulations that facilitate staphylococcal acclimatization and persistence in varying terrestrial and host conditions, and their public health ramifications in these settings.
Collapse
|
39
|
Wang Z, Choi JE, Wu CC, Di Nardo A. Skin commensal bacteria Staphylococcus epidermidis promote survival of melanocytes bearing UVB-induced DNA damage, while bacteria Propionibacterium acnes inhibit survival of melanocytes by increasing apoptosis. PHOTODERMATOLOGY PHOTOIMMUNOLOGY & PHOTOMEDICINE 2018; 34:405-414. [PMID: 29974533 DOI: 10.1111/phpp.12411] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Revised: 06/21/2018] [Accepted: 06/29/2018] [Indexed: 11/29/2022]
Abstract
BACKGROUND/PURPOSE Skin commensal bacteria have been described to help orchestrate skin homeostasis, signaling through innate immunity pathways. This study for the first time aimed at studying the relationship between skin commensals and melanocytes after UVB exposure. METHODS An in vitro UVB radiation model with normal human epidermal melanocytes (NHMs) and skin commensal bacteria supernatant from Staphylococcus epidermidis and Propionibacterium acnes was established. Melanocytes DNA damage, cyclobutane pyrimidine dimers (CPD), and cellular proliferation marker Ki-67 were measured by ELISA and immunofluorescence staining. Cell apoptosis was assessed by flow cytometry and PCR array and RT-qPCR. RESULTS Normal human epidermal melanocytes are able to survive and proliferate while bearing DNA damage after UVB radiation. Skin commensal bacteria S. epidermidis and its by-product LTA promote melanocytes survival by inducing upregulation of TRAF1, CASP14, CASP5, and TP73. On the other hand, P. acnes can inhibit UVB-irradiated melanocytes survival by increasing apoptosis. CONCLUSION Our studies show different aspects of commensal activity on melanocytes during irradiation. The possible balance achieved by the different skin commensal can influence NHM potential to become cancer cells.
Collapse
Affiliation(s)
- Zhenping Wang
- Department of Dermatology, University of California, San Diego, California
| | - Jae-Eun Choi
- Department of Dermatology, University of California, San Diego, California
| | - Chia-Chi Wu
- Department of Dermatology, University of California, San Diego, California
| | - Anna Di Nardo
- Department of Dermatology, University of California, San Diego, California
| |
Collapse
|
40
|
Glinel K, Behrens A, Langer RS, Jaklenec A, Jonas AM. Nanofibrillar Patches of Commensal Skin Bacteria. Biomacromolecules 2018; 20:102-108. [DOI: 10.1021/acs.biomac.8b00735] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Karine Glinel
- Institute of Condensed Matter and Nanosciences, Université Catholique de Louvain, Croix du Sud 1/L7.04.02, Louvain-la-Neuve, 1348, Belgium
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 500 Main Street, Cambridge, Massachusetts 02139, United States
| | - Adam Behrens
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 500 Main Street, Cambridge, Massachusetts 02139, United States
| | - Robert S. Langer
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 500 Main Street, Cambridge, Massachusetts 02139, United States
| | - Ana Jaklenec
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 500 Main Street, Cambridge, Massachusetts 02139, United States
| | - Alain M. Jonas
- Institute of Condensed Matter and Nanosciences, Université Catholique de Louvain, Croix du Sud 1/L7.04.02, Louvain-la-Neuve, 1348, Belgium
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, 500 Main Street, Cambridge, Massachusetts 02139, United States
| |
Collapse
|
41
|
Kundu RV, Mhlaba JM, Rangel SM, Le Poole IC. The convergence theory for vitiligo: A reappraisal. Exp Dermatol 2018; 28:647-655. [PMID: 29704874 DOI: 10.1111/exd.13677] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/18/2018] [Indexed: 12/15/2022]
Abstract
Vitiligo is characterized by progressive loss of skin pigmentation. The search for aetiologic factors has led to the biochemical, the neurologic and the autoimmune theory. The convergence theory was then proposed several years ago to incorporate existing theories of vitiligo development into a single overview of vitiligo aetiology. The viewpoint that vitiligo is not caused only by predisposing mutations, or only by melanocytes responding to chemical/radiation exposure, or only by hyperreactive T cells, but rather results from a combination of aetiologic factors that impact melanocyte viability, has certainly stood the test of time. New findings have since informed the description of progressive depigmentation. Understanding the relative importance of such aetiologic factors combined with a careful selection of the most targetable pathways will continue to drive the next phase in vitiligo research: the development of effective therapeutics. In that arena, it is likewise important to acknowledge that pathways affected in some patients may not be altered in others. Taken together, the convergence theory continues to provide a comprehensive viewpoint of vitiligo aetiology. The theory serves to intertwine aetiologic pathways and will help to define pathways amenable to disease intervention in individual patients.
Collapse
Affiliation(s)
- Roopal V Kundu
- Department of Dermatology, Northwestern University, Chicago, IL, USA
| | - Julia M Mhlaba
- Department of Dermatology, Northwestern University, Chicago, IL, USA
| | | | - I Caroline Le Poole
- Department of Dermatology, Northwestern University, Chicago, IL, USA.,Department of Microbiology and Immunology, Northwestern University, Chicago, IL, USA
| |
Collapse
|
42
|
Patra V, Laoubi L, Nicolas JF, Vocanson M, Wolf P. A Perspective on the Interplay of Ultraviolet-Radiation, Skin Microbiome and Skin Resident Memory TCRαβ+ Cells. Front Med (Lausanne) 2018; 5:166. [PMID: 29900173 PMCID: PMC5988872 DOI: 10.3389/fmed.2018.00166] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Accepted: 05/11/2018] [Indexed: 12/21/2022] Open
Abstract
The human skin is known to be inhabited by diverse microbes, including bacteria, fungi, viruses, archaea, and mites. This microbiome exerts a protective role against infections by promoting immune development and inhibiting pathogenic microbes to colonize skin. One of the factors having an intense effect on the skin and its resident microbes is ultraviolet-radiation (UV-R). UV-R can promote or inhibit the growth of microbes on the skin and modulate the immune system which can be either favorable or harmful. Among potential UV-R targets, skin resident memory T cells (TRM) stand as well positioned immune cells at the forefront within the skin. Both CD4+ or CD8+ αβ TRM cells residing permanently in peripheral tissues have been shown to play prominent roles in providing accelerated and long-lived specific immunity, tissue homeostasis, wound repair. Nevertheless, their response upon UV-R exposure or signals from microbiome are poorly understood compared to resident TCRγδ cells. Skin TRM survive for long periods of time and are exposed to innumerable antigens during lifetime. The interplay of TRM with skin residing microbes may be crucial in pathophysiology of various diseases including psoriasis, atopic dermatitis and polymorphic light eruption. In this article, we share our perspective about how UV-R may directly shape the persistence, phenotype, specificity, and function of skin TRM; and moreover, whether UV-R alters barrier function, leading to microbial-specific skin TRM, disrupting the healthy balance between skin microbiome and skin immune cells, and resulting in chronic inflammation and diseased skin.
Collapse
Affiliation(s)
- VijayKumar Patra
- Centre International de Recherche en Infectiologie, Institut National de la Santé et de la Recherche Médicale, U1111, Université Claude Bernard Lyon 1, Centre National de la Recherche Scientifique, UMR5308, Ecole Normale Supérieure de Lyon, Université de Lyon, Lyon, France.,Center for Medical Research, Medical University of Graz, Graz, Austria.,Research Unit for Photodermatology, Department of Dermatology, Medical University of Graz, Graz, Austria
| | - Léo Laoubi
- Centre International de Recherche en Infectiologie, Institut National de la Santé et de la Recherche Médicale, U1111, Université Claude Bernard Lyon 1, Centre National de la Recherche Scientifique, UMR5308, Ecole Normale Supérieure de Lyon, Université de Lyon, Lyon, France
| | - Jean-François Nicolas
- Centre International de Recherche en Infectiologie, Institut National de la Santé et de la Recherche Médicale, U1111, Université Claude Bernard Lyon 1, Centre National de la Recherche Scientifique, UMR5308, Ecole Normale Supérieure de Lyon, Université de Lyon, Lyon, France.,Allergy and Clinical Immunology Department, Lyon Sud University Hospital, Pierre-Bénite, France
| | - Marc Vocanson
- Centre International de Recherche en Infectiologie, Institut National de la Santé et de la Recherche Médicale, U1111, Université Claude Bernard Lyon 1, Centre National de la Recherche Scientifique, UMR5308, Ecole Normale Supérieure de Lyon, Université de Lyon, Lyon, France
| | - Peter Wolf
- Research Unit for Photodermatology, Department of Dermatology, Medical University of Graz, Graz, Austria
| |
Collapse
|
43
|
Jonas AM, Glinel K, Behrens A, Anselmo AC, Langer RS, Jaklenec A. Controlling the Growth of Staphylococcus epidermidis by Layer-By-Layer Encapsulation. ACS APPLIED MATERIALS & INTERFACES 2018; 10:16250-16259. [PMID: 29693369 DOI: 10.1021/acsami.8b01988] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Commensal skin bacteria such as Staphylococcus epidermidis are currently being considered as possible components in skin-care and skin-health products. However, considering the potentially adverse effects of commensal skin bacteria if left free to proliferate, it is crucial to develop methodologies that are capable of maintaining bacteria viability while controlling their proliferation. Here, we encapsulate S. epidermidis in shells of increasing thickness using layer-by-layer assembly, with either a pair of synthetic polyelectrolytes or a pair of oppositely charged polysaccharides. We study the viability of the cells and their delay of growth depending on the composition of the shell, its thickness, the charge of the last deposited layer, and the degree of aggregation of the bacteria which is varied using different coating procedures-among which is a new scalable process that easily leads to large amounts of nonaggregated bacteria. We demonstrate that the growth of bacteria is not controlled by the mechanical properties of the shell but by the bacteriostatic effect of the polyelectrolyte complex, which depends on the shell thickness and charge of its outmost layer, and involves the diffusion of unpaired amine sites through the shell. The lag times of growth are sufficient to prevent proliferation for daily topical applications.
Collapse
Affiliation(s)
- Alain M Jonas
- Institute of Condensed Matter and Nanosciences , Université catholique de Louvain , Croix du Sud 1/L7.04.02 , Louvain-la-Neuve 1348 , Belgium
- David H. Koch Institute for Integrative Cancer Research , Massachusetts Institute of Technology , 500 Main Street , Cambridge , Massachusetts 02139 , United States
| | - Karine Glinel
- Institute of Condensed Matter and Nanosciences , Université catholique de Louvain , Croix du Sud 1/L7.04.02 , Louvain-la-Neuve 1348 , Belgium
- David H. Koch Institute for Integrative Cancer Research , Massachusetts Institute of Technology , 500 Main Street , Cambridge , Massachusetts 02139 , United States
| | - Adam Behrens
- David H. Koch Institute for Integrative Cancer Research , Massachusetts Institute of Technology , 500 Main Street , Cambridge , Massachusetts 02139 , United States
| | - Aaron C Anselmo
- David H. Koch Institute for Integrative Cancer Research , Massachusetts Institute of Technology , 500 Main Street , Cambridge , Massachusetts 02139 , United States
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy , University of North Carolina at Chapel Hill , Chapel Hill , North Carolina 27599 , United States
| | - Robert S Langer
- David H. Koch Institute for Integrative Cancer Research , Massachusetts Institute of Technology , 500 Main Street , Cambridge , Massachusetts 02139 , United States
| | - Ana Jaklenec
- David H. Koch Institute for Integrative Cancer Research , Massachusetts Institute of Technology , 500 Main Street , Cambridge , Massachusetts 02139 , United States
| |
Collapse
|
44
|
Kabashima K, Biedermann T. A new era for translational atopic dermatitis research and management. Exp Dermatol 2018; 27:313-317. [DOI: 10.1111/exd.13546] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Affiliation(s)
- Kenji Kabashima
- Faculty of Medicine; Department of Dermatology; Graduate School of Medicine; Kyoto University; Kyoto Japan
| | - Tilo Biedermann
- Department of Dermatology and Allergy; Technical University of Munich; Munich Germany
- Clinical Unit Allergology; Helmholtz Zentrum München; German Research Center for Environmental Health (GmbH); Germany
| |
Collapse
|
45
|
Kim BE, Leung DYM. Significance of Skin Barrier Dysfunction in Atopic Dermatitis. ALLERGY, ASTHMA & IMMUNOLOGY RESEARCH 2018; 10:207-215. [PMID: 29676067 PMCID: PMC5911439 DOI: 10.4168/aair.2018.10.3.207] [Citation(s) in RCA: 224] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Revised: 10/31/2017] [Accepted: 11/08/2017] [Indexed: 12/13/2022]
Abstract
The epidermis contains epithelial cells, immune cells, and microbes which provides a physical and functional barrier to the protection of human skin. It plays critical roles in preventing environmental allergen penetration into the human body and responsing to microbial pathogens. Atopic dermatitis (AD) is the most common, complex chronic inflammatory skin disease. Skin barrier dysfunction is the initial step in the development of AD. Multiple factors, including immune dysregulation, filaggrin mutations, deficiency of antimicrobial peptides, and skin dysbiosis contribute to skin barrier defects. In the initial phase of AD, treatment with moisturizers improves skin barrier function and prevents the development of AD. With the progression of AD, effective topical and systemic therapies are needed to reduce immune pathway activation and general inflammation. Targeted microbiome therapy is also being developed to correct skin dysbiosis associated with AD. Improved identification and characterization of AD phenotypes and endotypes are required to optimize the precision medicine approach to AD.
Collapse
Affiliation(s)
- Byung Eui Kim
- Department of Pediatrics, National Jewish Health, Denver, CO, USA
| | - Donald Y M Leung
- Department of Pediatrics, National Jewish Health, Denver, CO, USA.
| |
Collapse
|
46
|
Czarnowicki T, Krueger JG, Guttman-Yassky E. Novel concepts of prevention and treatment of atopic dermatitis through barrier and immune manipulations with implications for the atopic march. J Allergy Clin Immunol 2017; 139:1723-1734. [PMID: 28583445 DOI: 10.1016/j.jaci.2017.04.004] [Citation(s) in RCA: 160] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Revised: 04/07/2017] [Accepted: 04/13/2017] [Indexed: 12/16/2022]
Abstract
Skin barrier abnormalities have been suggested to play an essential role in initiation of early atopic dermatitis (AD). Antigen penetration through a compromised barrier likely leads to increased innate immune responses, antigen-presenting cell stimulation, and priming of overt cutaneous disease. In a TH2-promoting environment, T-cell/B-cell interactions occurring in regional lymph nodes lead to excessive IgE switch. Concurrent redistribution of memory T cells into the circulation not only leads to exacerbation of AD through T-cell skin infiltration but also spreads beyond the skin to initiate the atopic march, which includes food allergy, asthma, and allergic rhinitis. Possible primary interventions to prevent AD are focusing on improving skin barrier integrity, including supplementing barrier function with moisturizers. As for secondary prophylaxis in children with established AD, this can be stratified into prevention of disease exacerbations by using proactive approaches (with either topical corticosteroids or topical calcineurin inhibitors) in mild AD cases or the prevention of other atopic disorders that will probably mandate systemic immunosuppression in severe AD cases.
Collapse
Affiliation(s)
- Tali Czarnowicki
- Department of Dermatology and the Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY; Laboratory for Investigative Dermatology, Rockefeller University, New York, NY
| | - James G Krueger
- Department of Dermatology and the Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY; Laboratory for Investigative Dermatology, Rockefeller University, New York, NY
| | - Emma Guttman-Yassky
- Department of Dermatology and the Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY; Laboratory for Investigative Dermatology, Rockefeller University, New York, NY.
| |
Collapse
|
47
|
Miodovnik M, Künstner A, Langan EA, Zillikens D, Gläser R, Sprecher E, Baines JF, Schmidt E, Ibrahim SM. A distinct cutaneous microbiota profile in autoimmune bullous disease patients. Exp Dermatol 2017; 26:1221-1227. [DOI: 10.1111/exd.13357] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/04/2017] [Indexed: 12/12/2022]
Affiliation(s)
- Mor Miodovnik
- Department of Dermatology; Tel Aviv Sourasky Medical Center; Tel Aviv Israel
| | - Axel Künstner
- Lübeck Institute of Experimental Dermatology (LIED); University of Lübeck; Lübeck Germany
- Max Planck Institute for Evolutionary Biology; Plön Germany
| | - Ewan A. Langan
- Institute of Medical Microbiology und Hygiene; University of Lübeck; Lübeck Germany
- Department of Dermatological Science; Division of Musculoskeletal & Dermatological Sciences; University of Manchester; Manchester UK
- Department of Dermatology; University of Lübeck; Lübeck Germany
| | - Detlef Zillikens
- Lübeck Institute of Experimental Dermatology (LIED); University of Lübeck; Lübeck Germany
- Department of Dermatology; University of Lübeck; Lübeck Germany
| | - Regine Gläser
- Department of Dermatology; Christian-Albrechts-University of Kiel; Kiel Germany
| | - Eli Sprecher
- Department of Dermatology; Tel Aviv Sourasky Medical Center; Tel Aviv Israel
| | - John F. Baines
- Max Planck Institute for Evolutionary Biology; Plön Germany
- Institute for Experimental Medicine; Christian-Albrechts-University of Kiel; Kiel Germany
| | - Enno Schmidt
- Lübeck Institute of Experimental Dermatology (LIED); University of Lübeck; Lübeck Germany
- Department of Dermatology; University of Lübeck; Lübeck Germany
| | - Saleh M. Ibrahim
- Lübeck Institute of Experimental Dermatology (LIED); University of Lübeck; Lübeck Germany
- Department of Dermatology; University of Lübeck; Lübeck Germany
| |
Collapse
|
48
|
Georgountzou A, Papadopoulos NG. Postnatal Innate Immune Development: From Birth to Adulthood. Front Immunol 2017; 8:957. [PMID: 28848557 PMCID: PMC5554489 DOI: 10.3389/fimmu.2017.00957] [Citation(s) in RCA: 120] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Accepted: 07/26/2017] [Indexed: 12/20/2022] Open
Abstract
It is well established that adaptive immune responses are deficient in early life, contributing to increased mortality and morbidity. The developmental trajectories of different components of innate immunity are only recently being explored. Individual molecules, cells, or pathways of innate recognition and signaling, within different compartments/anatomical sites, demonstrate variable maturation patterns. Despite some discrepancies among published data, valuable information is emerging, showing that the developmental pattern of cytokine responses during early life is age and toll-like receptor specific, and may be modified by genetic and environmental factors. Interestingly, specific environmental exposures have been linked both to innate function modifications and the occurrence of chronic inflammatory disorders, such as respiratory allergies. As these conditions are on the rise, our knowledge on innate immune development and its modulating factors needs to be expanded. Improved understanding of the sequence of events associated with disease onset and persistence will lead toward meaningful interventions. This review describes the state-of-the-art on normal postnatal innate immune ontogeny and highlights research areas that are currently explored or should be further addressed.
Collapse
Affiliation(s)
- Anastasia Georgountzou
- Allergy and Clinical Immunology Unit, 2nd Pediatric Clinic, National and Kapodistrian University of Athens, Athens, Greece
| | - Nikolaos G Papadopoulos
- Allergy and Clinical Immunology Unit, 2nd Pediatric Clinic, National and Kapodistrian University of Athens, Athens, Greece.,Division of Infection, Inflammation and Respiratory Medicine, The University of Manchester, Manchester, United Kingdom
| |
Collapse
|
49
|
The influence of the commensal microbiota on distal tumor-promoting inflammation. Semin Immunol 2017; 32:62-73. [PMID: 28687194 DOI: 10.1016/j.smim.2017.06.002] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Revised: 05/29/2017] [Accepted: 06/20/2017] [Indexed: 02/07/2023]
Abstract
Commensal microbes inhabit barrier surfaces, providing a first line of defense against invading pathogens, aiding in metabolic function of the host, and playing a vital role in immune development and function. Several recent studies have demonstrated that commensal microbes influence systemic immune function and homeostasis. For patients with extramucosal cancers, or cancers occurring distal to barrier surfaces, the role of commensal microbes in influencing tumor progression is beginning to be appreciated. Extrinsic factors such as chronic inflammation, antibiotics, and chemotherapy dysregulate commensal homeostasis and drive tumor-promoting systemic inflammation through a variety of mechanisms, including disruption of barrier function and bacterial translocation, release of soluble inflammatory mediators, and systemic changes in metabolic output. Conversely, it has also been demonstrated that certain immune therapies, immunogenic chemotherapies, and checkpoint inhibitors rely on the commensal microbiota to facilitate anti-tumor immune responses. Thus, it is evident that the mechanisms associated with commensal microbe facilitation of both pro- and anti-tumor immune responses are context dependent and rely upon a variety of factors present within the tumor microenvironment and systemic periphery. The goal of this review is to highlight the various contexts during which commensal microbes orchestrate systemic immune function with a focus on describing possible scenarios where the loss of microbial homeostasis enhances tumor progression.
Collapse
|
50
|
Advances in atopic dermatitis and urticarial in 2016. J Allergy Clin Immunol 2017; 140:369-376. [PMID: 28652155 DOI: 10.1016/j.jaci.2017.06.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Revised: 05/15/2017] [Accepted: 06/15/2017] [Indexed: 01/07/2023]
Abstract
This review highlights recent key advances in the pathology and therapies of inflammatory skin diseases, focusing on atopic dermatitis (AD) and chronic spontaneous urticaria (CSU). Regarding AD, transcriptomic analysis with human samples revealed different immune profiles between childhood and adult AD. Phase III clinical trials of dupilumab, an anti-IL-4 receptor α antibody, in the treatment of AD have successfully finished, and dupilumab will appear in clinical practice as the first biologic for AD in 2017. In addition, a novel biologic that targets IL-31 shows promising results in a phase II trial. As for the skin microbiome study, novel insights into the mechanisms of microbial dysbiosis, such as colonization of Staphylococcus aureus, a common feature of AD, were proposed. Regarding CSU, autoreactive CD4+ T cells that react to FcεRI were discovered, which might contribute to the development of CSU. These findings provide new insights into the pathogenesis of AD and CSU and will lead to more specific and personalized treatments.
Collapse
|