1
|
Chen H, Chen Y, Zhou Y, Cao S, Lu J, Han L, Worzfeld T, Krutmann J, Wang J, Xia J. Optimizing Skin Surface Metabolomics: A Comprehensive Evaluation of Sampling Methods, Extraction Solvents, and Analytical Techniques. J Invest Dermatol 2025; 145:1166-1179. [PMID: 39306031 DOI: 10.1016/j.jid.2024.08.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 08/21/2024] [Accepted: 08/22/2024] [Indexed: 11/04/2024]
Abstract
Characterizing the metabolite fingerprint from the skin surface provides invaluable insights into skin biology and microbe-host interactions. To ensure data accuracy and reproducibility, it is essential to develop standard operating procedures for skin surface metabolomics. However, there is a notable lack of studies in this area. In this study, we thoroughly evaluated different sampling materials, extraction solvents, taping methods (frequency and number of tapes), and analytical techniques to optimize skin surface metabolomics. Our results showed that the combination of D-Squame D100 tape with a methyl tert-butyl ether/methanol extractant is optimal for skin surface lipidomics. Performing the skin-taping procedure 5 times with 1 tape yields sufficient biomass for lipid analysis, whereas the optimal taping procedure varies for water-soluble compounds. In addition, our study identified associations among the skin surface metabolites, some of which potentially underlie the formation of microbial cutotypes and offer insights into host-microbe interactions.
Collapse
Affiliation(s)
- Huizhen Chen
- Human Phenome Institute, School of Life Sciences, Fudan University, Shanghai, China; Greater Bay Area Institute of Precision Medicine (Guangzhou), School of Life Sciences, Fudan University, Shanghai, China
| | - Yu Chen
- Wuhan Metware Biotechnology, Wuhan, China
| | - Yi Zhou
- Institute of Dermatology and Department of Dermatology, First Affiliated Hospital, Anhui Medical University, Hefei, China; Key Laboratory of Dermatology, Anhui Medical University, Ministry of Education, Hefei, China
| | - Shensong Cao
- Human Phenome Institute, School of Life Sciences, Fudan University, Shanghai, China
| | - Jing Lu
- Greater Bay Area Institute of Precision Medicine (Guangzhou), School of Life Sciences, Fudan University, Shanghai, China
| | - Lianyi Han
- Greater Bay Area Institute of Precision Medicine (Guangzhou), School of Life Sciences, Fudan University, Shanghai, China; Department of Dermatology, Huashan Hospital, Fudan University, Shanghai, China
| | - Thomas Worzfeld
- Institute of Pharmacology, University of Marburg, Marburg, Germany
| | - Jean Krutmann
- Human Phenome Institute, School of Life Sciences, Fudan University, Shanghai, China; IUF Leibniz Research Institute for Environmental Medicine, Düsseldorf, Germany
| | - Jiucun Wang
- Human Phenome Institute, School of Life Sciences, Fudan University, Shanghai, China; Department of Dermatology, Huashan Hospital, Fudan University, Shanghai, China; Research Unit of Dissecting the Population Genetics and Developing New Technologies for Treatment and Prevention of Skin Phenotypes and Dermatological Diseases, Chinese Academy of Medical Sciences, Shanghai, China.
| | - Jingjing Xia
- Greater Bay Area Institute of Precision Medicine (Guangzhou), School of Life Sciences, Fudan University, Shanghai, China.
| |
Collapse
|
2
|
Olah P, Reuvers N, Radai Z, Varadi A, van Lierop A, Wachtmeister T, Plante S, Chaskar P, Thomas C, Julia V, Alenius H, Homey B. Microbe-Host Interaction in Rosacea and its Modulation Through Topical Ivermectin. J Invest Dermatol 2025:S0022-202X(25)00398-7. [PMID: 40220854 DOI: 10.1016/j.jid.2025.03.031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Revised: 03/05/2025] [Accepted: 03/14/2025] [Indexed: 04/14/2025]
Abstract
Rosacea is characterized by inflammatory lesions, often accompanied by an increased density of the common skin mite Demodex folliculorum. Although rosacea shows a high prevalence and significantly affects the quality of life of patients, the underlying mechanisms, especially the role of cutaneous dysbiosis are largely unknown. Hence, we aimed to systematically characterize disease severity of rosacea patients in the context of mite density, the cutaneous microbiome and the host's transcriptome before and after 30 days of topical 1% ivermectin cream treatment. At day 30, a marked decrease in mite density was observed in 87.5% of patients. At day 0, distinct microbial community changes included the decrease in Cutibacterium acnes abundance, while Staphylococcus epidermidis colonization increased when compared to healthy volunteers. Interestingly, the insect symbiont Snodgrassella alvi was recovered from a highly Demodex-colonized patient and eradicated by day 30 of treatment. Although topical ivermectin did not affect bacterial dysbiosis, the host's transcriptome significantly normalized and an "ivermectin transcriptomic signature" was defined. Findings of the present study support that rosacea lesions are associated with dysbiosis. However, improvement of clinical signs during topical ivermectin is not associated with normalization of the bacterial microbiome, but rather a decrease of transcriptomic dysregulation and mite density.
Collapse
Affiliation(s)
- Peter Olah
- Department of Dermatology, University Hospital Düsseldorf, Medical Faculty, Heinrich-Heine-University Duesseldorf, Duesseldorf, Germany
| | - Nina Reuvers
- Department of Dermatology, University Hospital Düsseldorf, Medical Faculty, Heinrich-Heine-University Duesseldorf, Duesseldorf, Germany
| | - Zoltan Radai
- Department of Dermatology, University Hospital Düsseldorf, Medical Faculty, Heinrich-Heine-University Duesseldorf, Duesseldorf, Germany
| | - Alex Varadi
- Molecular Medicine Research Group, Szentágothai Research Centre, University of Pécs, Pécs, Hungary
| | - Anke van Lierop
- Department of Dermatology, University Hospital Düsseldorf, Medical Faculty, Heinrich-Heine-University Duesseldorf, Duesseldorf, Germany
| | - Thorsten Wachtmeister
- Biological and Medical Research Center (BMFZ), Medical Faculty, Heinrich-Heine-University, Duesseldorf, Duesseldorf, Germany
| | - Sandra Plante
- Biological and Medical Research Center (BMFZ), Medical Faculty, Heinrich-Heine-University, Duesseldorf, Duesseldorf, Germany
| | | | - Carla Thomas
- Department of Dermatology, University Hospital Düsseldorf, Medical Faculty, Heinrich-Heine-University Duesseldorf, Duesseldorf, Germany
| | | | - Harri Alenius
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Bernhard Homey
- Department of Dermatology, University Hospital Düsseldorf, Medical Faculty, Heinrich-Heine-University Duesseldorf, Duesseldorf, Germany.
| |
Collapse
|
3
|
Kang S, Jeon S, Baek H, Hwang S, Kim S, Youn SH, Kim JW, Jun SH, Kang NG. Lactobacillus-derived artificial extracellular vesicles for skin rejuvenation and prevention of photo-aging. Biomater Sci 2025; 13:2026-2035. [PMID: 40013489 DOI: 10.1039/d4bm01644k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/28/2025]
Abstract
Extracellular vesicles (EVs) are small membrane-bound sacs released by cells that play crucial roles in intercellular communication. They transport biomolecules between cells and have both diagnostic and therapeutic potential. Artificial EVs, designed to mimic natural EVs, have been developed using various methods. In this study, Lactobacillus plantarum was used to create Lactobacillus-derived artificial EVs (LAEs) for skin rejuvenation and anti-aging. LAEs demonstrated monodispersity and effectively improved adverse gene expression and wound healing in fibroblasts. They also modulated aging-related genes and improved skin conditions in humans. Their simplicity, promptness, and lack of animal-derived sources make LAEs a promising alternative to natural EVs. LAEs have the potential to overcome the technical limitations of artificial EVs and advance EVs or exosome-based technologies for comprehensive skin rejuvenation.
Collapse
Affiliation(s)
- Seongsu Kang
- LG Household and Health Care R&D Center, Seoul 07795, Republic of Korea.
| | - Saetbyeol Jeon
- School of Chemical Engineering, Sungkyunkwan University, Seobu-ro, Jangan-gu, Suwon, Gyeonggi-do 16419, Republic of Korea.
| | - Hwira Baek
- School of Chemical Engineering, Sungkyunkwan University, Seobu-ro, Jangan-gu, Suwon, Gyeonggi-do 16419, Republic of Korea.
| | - Sunghwan Hwang
- LG Household and Health Care R&D Center, Seoul 07795, Republic of Korea.
| | - Seulgi Kim
- School of Chemical Engineering, Sungkyunkwan University, Seobu-ro, Jangan-gu, Suwon, Gyeonggi-do 16419, Republic of Korea.
| | - Sung Hun Youn
- LG Household and Health Care R&D Center, Seoul 07795, Republic of Korea.
| | - Jin Woong Kim
- School of Chemical Engineering, Sungkyunkwan University, Seobu-ro, Jangan-gu, Suwon, Gyeonggi-do 16419, Republic of Korea.
| | - Seung-Hyun Jun
- LG Household and Health Care R&D Center, Seoul 07795, Republic of Korea.
| | - Nae-Gyu Kang
- LG Household and Health Care R&D Center, Seoul 07795, Republic of Korea.
| |
Collapse
|
4
|
Ćorović M, Petrov Ivanković A, Milivojević A, Pfeffer K, Homey B, Jansen PAM, Zeeuwen PLJM, van den Bogaard EH, Bezbradica D. Investigating the Effect of Enzymatically-Derived Blackcurrant Extract on Skin Staphylococci Using an In Vitro Human Stratum Corneum Model. Pharmaceutics 2025; 17:487. [PMID: 40284482 PMCID: PMC12030025 DOI: 10.3390/pharmaceutics17040487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2025] [Revised: 03/31/2025] [Accepted: 04/03/2025] [Indexed: 04/29/2025] Open
Abstract
Background/Objectives: Numerous intrinsic and extrinsic stressors can disrupt the balance of the skin microbiome, leading to the development of various skin diseases. It has been proven that coagulase-negative staphylococci (CoNS) are important commensals for maintaining skin microbiome homeostasis and fighting cutaneous pathogens such as Staphylococcus aureus (S. aureus). Here, we examined the influence of polyphenol-rich enzymatic blackcurrant extract (EBCE) on pathogenic coagulase-positive S. aureus strains and beneficial CoNS, like Staphylococcus epidermidis (S. epidermidis), to explore its potential for rebalancing the skin microbiota. Methods: The polyphenol profile of EBCE was determined by ultra-high-pressure liquid chromatography-tandem mass spectrometry. Microwell plate assays were employed to study the effect of EBCE on five S. aureus strains isolated from the skin of atopic dermatitis patients. An in vitro human stratum corneum model was used to test its effect on mixed bacterial cultures. Results: EBCE inhibited the growth of all tested S. aureus strains by 80-100% at the highest tested concentration after 7 h. No microbial growth was observed at the highest tested EBCE concentration using the stratum corneum model inoculated with one selected pathogen (S. aureus SA-DUS-017) and one commensal laboratory strain (S. epidermidis DSM 20044). The lowest tested concentration did not interfere with S. aureus growth but strongly stimulated the growth of S. epidermidis (~300-fold colony forming unit increase). In addition, low EBCE concentrations strongly stimulated CoNS growth in microbiome samples taken from the armpits of healthy volunteers that were spiked with S. aureus SA-DUS-017. Conclusions: These preclinical data support further testing of EBCE-enriched topical preparations as potential cutaneous prebiotics in human studies.
Collapse
Affiliation(s)
- Marija Ćorović
- Faculty of Technology and Metallurgy, University of Belgrade, Karnegijeva 4, 11000 Belgrade, Serbia; (A.M.); (D.B.)
| | - Anja Petrov Ivanković
- Innovation Center, Faculty of Technology and Metallurgy, Karnegijeva 4, 11000 Belgrade, Serbia;
| | - Ana Milivojević
- Faculty of Technology and Metallurgy, University of Belgrade, Karnegijeva 4, 11000 Belgrade, Serbia; (A.M.); (D.B.)
| | - Klaus Pfeffer
- Department of Microbiology, University Hospital Düsseldorf, Medical Faculty, Heinrich-Heine-University Düsseldorf, 40225 Düsseldorf, Germany;
| | - Bernhard Homey
- Department of Dermatology, University Hospital Düsseldorf, Medical Faculty, Heinrich-Heine-University Düsseldorf, 40225 Düsseldorf, Germany;
| | - Patrick A. M. Jansen
- Department of Dermatology, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands; (P.A.M.J.); (P.L.J.M.Z.); (E.H.v.d.B.)
| | - Patrick L. J. M. Zeeuwen
- Department of Dermatology, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands; (P.A.M.J.); (P.L.J.M.Z.); (E.H.v.d.B.)
| | - Ellen H. van den Bogaard
- Department of Dermatology, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands; (P.A.M.J.); (P.L.J.M.Z.); (E.H.v.d.B.)
| | - Dejan Bezbradica
- Faculty of Technology and Metallurgy, University of Belgrade, Karnegijeva 4, 11000 Belgrade, Serbia; (A.M.); (D.B.)
| |
Collapse
|
5
|
Sun L, Wang Q, Huang J, Wang H, Yu Z. Disrupting the balance: how acne duration impacts skin microbiota assembly processes. Microbiol Spectr 2025; 13:e0260324. [PMID: 39992142 PMCID: PMC11960176 DOI: 10.1128/spectrum.02603-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Accepted: 01/28/2025] [Indexed: 02/25/2025] Open
Abstract
Growing interest in the impact of microbial balance on health has driven studies on the ecological processes shaping the skin microbiota. Skin diseases, which alter the skin's local environment, can disrupt the microbial structure and interact with the disease itself. However, research on microbial assembly in diseased skin remains limited. In this study, we applied ecological models to characterize the processes shaping the skin microbiota in acne patients, considering the impact of disease duration on both skin pores and surfaces using bacterial amplicon sequencing. Our results revealed a significant shift in microbial diversity on the skin surface of patients with long-term acne. Further microbial community analyses showed a transition in ecological processes from healthy to diseased skin. Microbial communities on the skin surfaces of healthy controls and individuals with short-duration acne were primarily driven by heterogeneous selection, whereas microbial drift dominated the assembly process in the long-duration groups. Using the Sloan neutral model, we classified amplicon sequence variants (ASVs) into high-effect and low-effect groups based on relative abundance and sample occurrence. High-effect ASVs, likely exerting a greater ecological influence, were predominantly represented by Cutibacterium across all acne-affected skin groups, while Staphylococcus became enriched among high-effect ASVs in patients with long-term acne. Functional profiling further demonstrated that high-effect ASVs were significantly enriched in motility-related pathways. Additionally, we observed a reduction in microbial network complexity on skin surfaces as disease duration increased. Overall, the ecological dynamics of skin microbial communities may offer valuable insights into the mechanisms underlying disease onset and persistence.IMPORTANCEThe skin microbiota plays a critical role in acne development, yet the processes governing microbial assembly during acne progression remain poorly understood. Previous studies predominantly focused on factors such as acne severity, location, and duration in relation to skin microbial structure, with little attention given to the ecological mechanisms shaping the communities. In this study, we applied ecological models to investigate the processes influencing microbial assembly of skin microbiota in acne patients with varying disease durations and examined functions of ecologically important non-neutral amplicon sequence variants (ASVs). Our findings reveal a transition in ecological processes from deterministic to neutral processes as acne duration increased, with non-neutral ASVs potentially contributing to acne pathogenicity and persistence. These insights contribute to a deeper understanding of the ecological dynamics underlying acne and indicate that targeting these non-neutral ASVs or their associated functions may serve as the basis for future therapeutic strategies.
Collapse
Affiliation(s)
- Lang Sun
- Department of Microbiology, Human Microbiome and Health Group, School of Basic Medical Science, Central South University, Changsha, Hunan, China
| | - Qingqun Wang
- Department of Microbiology, Human Microbiome and Health Group, School of Basic Medical Science, Central South University, Changsha, Hunan, China
| | - Jing Huang
- Department of Parasitology, School of Basic Medical Science, Central South University, Changsha, Hunan, China
| | - Huan Wang
- Department of Dermatology, The Fourth Hospital of Changsha, Changsha, China
| | - Zheng Yu
- Department of Microbiology, Human Microbiome and Health Group, School of Basic Medical Science, Central South University, Changsha, Hunan, China
| |
Collapse
|
6
|
Fukuda K, Ito Y, Amagai M. Barrier Integrity and Immunity: Exploring the Cutaneous Front Line in Health and Disease. Annu Rev Immunol 2025; 43:219-252. [PMID: 40279307 DOI: 10.1146/annurev-immunol-082323-030832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/27/2025]
Abstract
Immune responses are influenced by not only immune cells but also the tissue microenvironment where these cells reside. Recent advancements in understanding the underlying molecular mechanisms and structures of the epidermal tight junctions (TJs) and stratum corneum (SC) have significantly enhanced our knowledge of skin barrier functions. TJs, located in the granular layer of the epidermis, are crucial boundary elements in the differentiation process, particularly in the transition from living cells to dead cells. The SC forms from dead keratinocytes via corneoptosis and features three distinct pH zones critical for barrier function and homeostasis. Additionally, the SC-skin microbiota interactions are crucial for modulating immune responses and protecting against pathogens. In this review, we explore how these components contribute both to healthy and disease states. By targeting the skin barrier in therapeutic strategies, we can enhance its integrity, modulate immune responses, and ultimately improve outcomes for patients with inflammatory skin conditions.
Collapse
Affiliation(s)
- Keitaro Fukuda
- Department of Dermatology, Keio University School of Medicine, Tokyo, Japan;
- Laboratory for Skin Homeostasis, RIKEN Center for Integrative Medical Sciences, Kanagawa, Japan;
| | - Yoshihiro Ito
- Department of Dermatology, Keio University School of Medicine, Tokyo, Japan;
| | - Masayuki Amagai
- Department of Dermatology, Keio University School of Medicine, Tokyo, Japan;
- Laboratory for Skin Homeostasis, RIKEN Center for Integrative Medical Sciences, Kanagawa, Japan;
| |
Collapse
|
7
|
Cha J, Kim TG, Ryu JH. Conversation between skin microbiota and the host: from early life to adulthood. Exp Mol Med 2025; 57:703-713. [PMID: 40164684 DOI: 10.1038/s12276-025-01427-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 12/29/2024] [Accepted: 01/05/2025] [Indexed: 04/02/2025] Open
Abstract
Host life is inextricably linked to commensal microbiota, which play a crucial role in maintaining homeostasis and immune activation. A diverse array of commensal microbiota on the skin interacts with the host, influencing the skin physiology in various ways. Early-life exposure to commensal microbiota has long-lasting effects, and disruption of the epidermal barrier or transient exposure to these microorganisms can lead to skin dysbiosis and inflammation. Several commensal skin microbiota have the potential to function as either commensals or pathogens, both influencing and being influenced by the pathogenesis of skin inflammatory diseases. Here we explore the impact of various commensal skin microbiota on the host and elucidate the interactions between skin microbiota and host systems. A deeper understanding of these interactions may open new avenues for developing effective strategies to address skin diseases.
Collapse
Affiliation(s)
- Jimin Cha
- Department of Biomedical Sciences, Yonsei University College of Medicine, Seoul, Republic of Korea
- Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Tae-Gyun Kim
- Department of Dermatology, Severance Hospital, Cutaneous Biology Research Institute, Yonsei University College of Medicine, Seoul, Republic of Korea
- Institute for Immunology and Immunological Diseases, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Ji-Hwan Ryu
- Department of Biomedical Sciences, Yonsei University College of Medicine, Seoul, Republic of Korea.
- Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
8
|
Lee HJ, Kim HS. Prurigo nodularis and the microbiome. Clin Dermatol 2025:S0738-081X(25)00090-2. [PMID: 40157400 DOI: 10.1016/j.clindermatol.2025.03.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/01/2025]
Abstract
Prurigo nodularis (PN) is a chronic skin condition that profoundly impacts quality of life. Histopathological studies of itchy hyperkeratotic nodules show dense infiltrates of T lymphocytes, mast cells, and eosinophils. A robust inflammatory response is implicated, coupled with key changes in neuronal plasticity that affect nerve fiber architecture and function. The microbial community in PN lesions exhibits a distinct composition, marked by decreased α-diversity and a prominent increase in Staphylococcus aureus (S aureus). This alteration appears to contribute to the disease's pathophysiology, causing further disruption of the skin barrier, immune dysregulation, and neuronal plasticity. There is ample evidence that virulence factors of S aureus promote Th2, Th17, and Th22 cytokine production, which are key to PN. In addition, S aureus V8 protease (Endoproteinase Glu-C) has recently been identified to trigger robust itch by activating protease-activated receptor 1 (PAR1) on sensory neurons. This review underscores the complex interplay between the altered microbiome and the itch-scratch cycle of PN, providing insights into potential therapeutics targeting the skin microbiome. A multidisciplinary approach is crucial for providing relief to individuals suffering from this skin condition.
Collapse
Affiliation(s)
- Hyun Ji Lee
- Department of Dermatology, Yeouido St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Hei Sung Kim
- Department of Dermatology, Incheon St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea.
| |
Collapse
|
9
|
Dong D, Wu Q, Wang Z, Xu P, Zhu T, Yang T, Gu Z, Zhang L, Guo Z. Multi-perspective analysis of skin microbiota in elderly eczema patients. PLoS One 2025; 20:e0318240. [PMID: 40146755 PMCID: PMC11949328 DOI: 10.1371/journal.pone.0318240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Accepted: 01/13/2025] [Indexed: 03/29/2025] Open
Abstract
Eczema is a common inflammatory skin disease in elderly people. It not only causes physical damage to elderly people but also seriously affects their mental health. The skin microbiota plays a vital role in the development of skin disease. However, relatively few studies have investigated the characteristics of the skin microbiota in elderly eczema patients. In this study, the differences in the composition of the skin microbiota between lesion sites and healthy sites, between exposed sites and unexposed sites, and between elderly and younger eczema sites were analyzed, aiming to characterize the skin microbiota in elderly eczema patients from multiple perspectives and provide a basis for clinical diagnosis and treatment. The results indicated that the species richness of elderly eczema patients was greater than that of younger eczema patients. There was no significant difference between groups at the phylum level. At the genus level, the abundance of Staphylococcus significantly increased in the lesion sites of the elderly group. Compared with the younger eczema group, the elderly eczema group had greater abundances of Paracoccus, Deinococcus_B, Kaistella, Escherichia_710834, and Chryseobacterium_796703. These findings indicated that more attention should be given to the roles of Kaistella and Streptococcus in elderly eczema patients because Kaistella was the only genus among the 20 most abundant genera that was closely related to the EASI scores. Moreover, correlation analysis suggested that many genera had a positive relationship with Streptococcus. The results provide basic microbiological data for physicians treating elderly patients with eczema.
Collapse
Affiliation(s)
- Dake Dong
- Department of Dermatology, Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Qianjie Wu
- Wuxi Medical College, Jiangnan University, Wuxi, China
| | - Zijun Wang
- School of Biotechnology, Jiangnan University, Wuxi, China
- National Engineering Research Center of Cereal Fermentation and Food Biomanufacturing, Jiangnan University, Wuxi, China
| | - Pengfei Xu
- School of Biotechnology, Jiangnan University, Wuxi, China
- National Engineering Research Center of Cereal Fermentation and Food Biomanufacturing, Jiangnan University, Wuxi, China
| | - Tianqing Zhu
- School of Biotechnology, Jiangnan University, Wuxi, China
- National Engineering Research Center of Cereal Fermentation and Food Biomanufacturing, Jiangnan University, Wuxi, China
| | - Ting Yang
- Wuxi Food Safety Inspection and Test Center, Technology Innovation Center of Special Food for State Market Regulation, Wuxi, China
| | - Zhenghua Gu
- School of Biotechnology, Jiangnan University, Wuxi, China
- National Engineering Research Center of Cereal Fermentation and Food Biomanufacturing, Jiangnan University, Wuxi, China
| | - Liang Zhang
- School of Biotechnology, Jiangnan University, Wuxi, China
- National Engineering Research Center of Cereal Fermentation and Food Biomanufacturing, Jiangnan University, Wuxi, China
| | - Zitao Guo
- School of Food and Biological Engineering, Jiangsu University, Zhenjiang, China
| |
Collapse
|
10
|
Periferakis A, Periferakis AT, Troumpata L, Periferakis K, Georgatos-Garcia S, Touriki G, Dragosloveanu CDM, Caruntu A, Savulescu-Fiedler I, Dragosloveanu S, Scheau AE, Badarau IA, Caruntu C, Scheau C. Pinosylvin: A Multifunctional Stilbenoid with Antimicrobial, Antioxidant, and Anti-Inflammatory Potential. Curr Issues Mol Biol 2025; 47:204. [PMID: 40136458 PMCID: PMC11941527 DOI: 10.3390/cimb47030204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2025] [Revised: 03/13/2025] [Accepted: 03/15/2025] [Indexed: 03/27/2025] Open
Abstract
Stilbenoids are a category of plant compounds exhibiting notable health-related benefits. After resveratrol, perhaps the most well-known stilbenoid is pinosylvin, a major phytochemical constituent of most plants characterised by the pine spines among others. Pinosylvin and its derivatives have been found to exert potent antibacterial and antifungal effects, while their antiparasitic and antiviral properties are still a subject of ongoing research. The antioxidant properties of pinosylvin are mostly based on its scavenging of free radicals, inhibition of iNOS and protein kinase C, and promotion of HO-1 expression. Its anti-inflammatory properties are based on a variety of mechanisms, such as COX-2 inhibition, NF-κB and TRPA1 activation inhibition, and reduction in IL-6 levels. Its anticancer properties are partly associated with its antioxidant and anti-inflammatory potential, although a number of other mechanisms are described, such as apoptosis induction and matrix metalloproteinase inhibition. A couple of experiments have also suggested a neuroprotective potential. A multitude of ethnomedical and ethnobotanical effects of pinosylvin-containing plants are reported, like antimicrobial, antioxidant, anti-inflammatory, hepatoprotective, and prokinetic actions; many of these are corroborated by recent research. The advent of novel methods of artificial pinosylvin synthesis may facilitate its mass production and adoption as a medical compound. Finally, pinosylvin may be a tool in promoting environmentally friendly pesticide and insecticide policies and be used in land remediation schemes.
Collapse
Affiliation(s)
- Argyrios Periferakis
- Department of Physiology, The “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Akadimia of Ancient Greek and Traditional Chinese Medicine, 16675 Athens, Greece
- Elkyda, Research & Education Centre of Charismatheia, 17675 Athens, Greece
| | - Aristodemos-Theodoros Periferakis
- Department of Physiology, The “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Elkyda, Research & Education Centre of Charismatheia, 17675 Athens, Greece
| | - Lamprini Troumpata
- Department of Physiology, The “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
| | - Konstantinos Periferakis
- Akadimia of Ancient Greek and Traditional Chinese Medicine, 16675 Athens, Greece
- Pan-Hellenic Organization of Educational Programs (P.O.E.P.), 17236 Athens, Greece
| | - Spyrangelos Georgatos-Garcia
- Tilburg Institute for Law, Technology, and Society (TILT), Tilburg University, 5037 DE Tilburg, The Netherlands
- Corvers Greece IKE, 15124 Athens, Greece
| | - Georgia Touriki
- Faculty of Law, Democritus University of Thrace, 69100 Komotini, Greece
| | - Christiana Diana Maria Dragosloveanu
- Department of Ophthalmology, Faculty of Dentistry, The “Carol Davila” University of Medicine and Pharmacy, 020021 Bucharest, Romania
- Department of Ophthalmology, Clinical Hospital for Ophthalmological Emergencies, 010464 Bucharest, Romania
| | - Ana Caruntu
- Department of Oral and Maxillofacial Surgery, “Carol Davila” Central Military Emergency Hospital, 010825 Bucharest, Romania
- Department of Oral and Maxillofacial Surgery, Faculty of Dental Medicine, Titu Maiorescu University, 031593 Bucharest, Romania
| | - Ilinca Savulescu-Fiedler
- Department of Internal Medicine, The “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Department of Internal Medicine and Cardiology, Coltea Clinical Hospital, 030167 Bucharest, Romania
| | - Serban Dragosloveanu
- Department of Orthopaedics and Traumatology, The “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Department of Orthopaedics, “Foisor” Clinical Hospital of Orthopaedics, Traumatology and Osteoarticular TB, 021382 Bucharest, Romania
| | - Andreea-Elena Scheau
- Department of Radiology and Medical Imaging, “Foisor” Clinical Hospital of Orthopaedics, Traumatology and Osteoarticular TB, 021382 Bucharest, Romania
| | - Ioana Anca Badarau
- Department of Physiology, The “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
| | - Constantin Caruntu
- Department of Physiology, The “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Department of Dermatology, “Prof. N.C. Paulescu” National Institute of Diabetes, Nutrition and Metabolic Diseases, 011233 Bucharest, Romania
| | - Cristian Scheau
- Department of Physiology, The “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Department of Radiology and Medical Imaging, “Foisor” Clinical Hospital of Orthopaedics, Traumatology and Osteoarticular TB, 021382 Bucharest, Romania
| |
Collapse
|
11
|
Huang F, Zhu X, Liang X. Research hotspots and trends of skin barrier in atopic dermatitis in the past 24-year: a bibliometric analysis. Front Med (Lausanne) 2025; 12:1539386. [PMID: 40144870 PMCID: PMC11936790 DOI: 10.3389/fmed.2025.1539386] [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: 12/04/2024] [Accepted: 02/28/2025] [Indexed: 03/28/2025] Open
Abstract
Background Atopic dermatitis (AD) is a chronic, pruritic, inflammatory skin condition that imposes significant psychological and economic burdens on patients due to its recurring nature. Its etiology is multifactorial, involving interactions between genetic predispositions and environmental factors. The skin barrier serves as both a mechanical and immunological defense, and its structural damage and functional impairments significantly contribute to the pathogenesis of AD. This study aims to explore the future prospects and developmental trends of the skin barrier in the context of AD through a bibliometric analysis. Objective To analyze the research status, hot spots and development trend of skin barrier in AD. Methods Relevant studies were extracted from the Web of Science database and screened by researchers, with bibliometric analysis conducted using VOSviewer, CiteSpace, and other tools. Results A total of 4,227 publications were identified over a 24-year research period. The United States is the leading contributor, with 1,263 publications, and demonstrates extensive collaboration with numerous countries. The journal with the highest number of publications is the Journal of Allergy and Clinical Immunology. The most prolific institutions is the University of California, San Francisco. Recent years have seen high citation intensity for keywords such as "dupilumab," "barrier dysfunction," and "gut microbiota". Conclusion The mechanism of the skin barrier in AD remains an area requiring ongoing research and analysis. Although significant progress has been achieved, future research will benefit from advancements in technology.
Collapse
Affiliation(s)
- Fangchang Huang
- The First Clinical College of Medicine, Guangdong Medical University, Zhanjiang, Guangdong, China
- Department of Dermatology, Maoming People’s Hospital, Maoming, Guangdong, China
| | - Xin Zhu
- The First Clinical College of Medicine, Guangdong Medical University, Zhanjiang, Guangdong, China
- Department of Dermatology, Maoming People’s Hospital, Maoming, Guangdong, China
| | - Xinglong Liang
- The First Clinical College of Medicine, Guangdong Medical University, Zhanjiang, Guangdong, China
- Department of Dermatology, Maoming People’s Hospital, Maoming, Guangdong, China
| |
Collapse
|
12
|
Yang L, Wang P, Gao T, Huang X, Lin Z, Sweren E, Li Y, Chen L, Alphonse MP, Zhang J, Wang G. Melatonin treatment increases skin microbiota-derived propionic acid to alleviate atopic dermatitis. J Allergy Clin Immunol 2025; 155:880-891.e9. [PMID: 39579877 DOI: 10.1016/j.jaci.2024.11.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 11/06/2024] [Accepted: 11/10/2024] [Indexed: 11/25/2024]
Abstract
BACKGROUND Melatonin has been reported to relieve the inflammatory symptoms and improve sleep disturbance in patients with atopic dermatitis (AD). Recent studies showed that melatonin produced beneficial effects by remodeling intestinal microbiota composition; however, whether the beneficial effects of melatonin in AD were mediated by the modulation of skin microbiota remains unclear. OBJECTIVE We sought to investigate the mechanism by which melatonin treatment-induced changes in the skin microbiota composition further alleviated AD. METHODS The changes in skin bacterial composition after melatonin treatment were detected by 16S-rRNA sequencing. Further mechanisms were explored in calcipotriol (MC903)-induced AD mice and HaCaT cells through skin microbiota transplantation, quantification detection of short-chain fatty acids, transcriptome and single-cell sequencing analysis, quantitative RT-PCR, Western blotting, and Cell Counting Kit-8 assay. RESULTS We demonstrated that melatonin reshaped the skin microbiota in AD mice. The transplantation of skin microbiota from melatonin-treated mice alleviated AD symptoms in mice. Skin microbiota-derived short-chain fatty acids, especially propionic acid, were increased in the skin of melatonin-treated AD mice, which further inhibited FABP5 expression to alleviate AD. Propionic acid also inhibited FABP5 expression in HaCaT cells, which was reversed by the treatment of GPR43 inhibitor GLPG0974. GLPG0974 also blocked the therapeutic effects of melatonin on AD mice. CONCLUSIONS Our study demonstrated that melatonin alleviates AD through the skin microbiota/propionic acid/GPR43/FABP5 axis, highlighting a novel role of melatonin as a modulator of skin microbiota to alleviate AD.
Collapse
Affiliation(s)
- Lan Yang
- Department of Plastic and Aesthetic Surgery, Nanfang Hospital of Southern Medical University, Guangzhou, China
| | - Piao Wang
- Department of Plastic and Aesthetic Surgery, Nanfang Hospital of Southern Medical University, Guangzhou, China
| | - Ting Gao
- Department of Rehabilitation, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Xinyu Huang
- Department of Plastic and Aesthetic Surgery, Nanfang Hospital of Southern Medical University, Guangzhou, China
| | - Zhen Lin
- Department of Plastic and Aesthetic Surgery, Nanfang Hospital of Southern Medical University, Guangzhou, China
| | - Evan Sweren
- University of Michigan Medical School, Ann Arbor, Mich
| | - Yue Li
- Department of Plastic and Aesthetic Surgery, Nanfang Hospital of Southern Medical University, Guangzhou, China
| | - Lu Chen
- Department of Plastic and Aesthetic Surgery, Nanfang Hospital of Southern Medical University, Guangzhou, China
| | - Martin P Alphonse
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, Md
| | - Jianglin Zhang
- Department of Dermatology, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, China.
| | - Gaofeng Wang
- Department of Plastic and Aesthetic Surgery, Nanfang Hospital of Southern Medical University, Guangzhou, China; Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, Md.
| |
Collapse
|
13
|
Kim HB, Alexander H, Um JY, Chung BY, Park CW, Flohr C, Kim HO. Skin Microbiome Dynamics in Atopic Dermatitis: Understanding Host-Microbiome Interactions. ALLERGY, ASTHMA & IMMUNOLOGY RESEARCH 2025; 17:165-180. [PMID: 40204503 PMCID: PMC11982640 DOI: 10.4168/aair.2025.17.2.165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 02/23/2025] [Accepted: 03/16/2025] [Indexed: 04/11/2025]
Abstract
Atopic dermatitis (AD) is a chronic inflammatory skin disorder affecting both children and adults, characterized by pruritus, eczematous lesions, and compromised skin barrier function. A key feature of AD is dysbiosis of the skin microbiome, marked by reduced microbial diversity and the overgrowth of Staphylococcus aureus in lesional skin. S. aureus exacerbates skin barrier dysfunction and immune dysregulation, leading to recurrent infections and disease flares. In contrast, commensal bacteria such as Staphylococcus epidermidis and Roseomonas mucosa may exert protective effects by inhibiting S. aureus colonization and modulating immune responses. Beyond microbial composition, microbial metabolites play a crucial role in AD pathophysiology. Short-chain fatty acids, indole derivatives, and other bacterial metabolites influence cutaneous immune responses, lipid metabolism, and skin barrier integrity. Altered metabolite profiles, including reduced levels of beneficial microbial metabolites, are associated with AD severity and disease progression. Notably, S. aureus overabundance correlates with disruption in lipid metabolism, further compromising the skin barrier. This review explores recent advances in understanding the relationship between microbial metabolites and AD pathogenesis and examines the therapeutic potential of microbiome-targeted interventions. Strategies such as probiotics, prebiotics, and topical microbiome transplantation aim to restore microbial diversity and rebalance metabolite production, ultimately improving clinical outcomes in AD patients. Future therapeutic approaches focusing on commensal-derived metabolites offer promising avenues for alleviating symptoms and modulating disease severity in AD.
Collapse
Affiliation(s)
- Han Bi Kim
- Department of Dermatology, Hallym University Kangnam Sacred Heart Hospital, Hallym University College of Medicine, Seoul, Korea
| | - Helen Alexander
- Dermatology & Population Health Science, St John's Institute of Dermatology, King's College London, London, United Kingdom
| | - Ji Young Um
- Department of Dermatology, Hallym University Kangnam Sacred Heart Hospital, Hallym University College of Medicine, Seoul, Korea
| | - Bo Young Chung
- Department of Dermatology, Hallym University Kangnam Sacred Heart Hospital, Hallym University College of Medicine, Seoul, Korea
| | - Chun Wook Park
- Department of Dermatology, Hallym University Kangnam Sacred Heart Hospital, Hallym University College of Medicine, Seoul, Korea
| | - Carsten Flohr
- Dermatology & Population Health Science, St John's Institute of Dermatology, King's College London, London, United Kingdom.
| | - Hye One Kim
- Department of Dermatology, Hallym University Kangnam Sacred Heart Hospital, Hallym University College of Medicine, Seoul, Korea.
| |
Collapse
|
14
|
Bi O, Caballero‐Lima D, Sikkink S, Westgate G, Kauser S, Elies J, Thornton MJ. Do Melanocytes Have a Role in Controlling Epidermal Bacterial Colonisation and the Skin Microbiome? Exp Dermatol 2025; 34:e70071. [PMID: 40051134 PMCID: PMC11885897 DOI: 10.1111/exd.70071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 02/09/2025] [Accepted: 02/17/2025] [Indexed: 03/10/2025]
Abstract
In addition to producing melanin to protect epidermal keratinocytes against DNA damage, melanocytes may have important roles in strengthening innate immunity against pathogens. We have developed a functional, pigmented, human full-thickness 3D skin equivalent to determine whether the presence of melanocytes impacts epidermal bacterial growth and regulates the expression of genes involved in the immune response. We introduced primary epidermal melanocytes to construct a 3-cell full-thickness skin equivalent with primary dermal fibroblasts and epidermal keratinocytes. Immunohistochemistry verified the appropriate ratio and spatial organisation of melanocytes. Alpha-MSH induced melanogenesis, confirming an appropriate physiological response. We compared this 3-cell skin equivalent with the 2-cell version without melanocytes in response to inoculation with 3 species of bacteria: Staphylococcus epidermidis, Corynebacterium striatum, and Cutibacterium acnes. There was a significant decrease in the colonisation of bacteria in the skin equivalents containing functional melanocytes. There was increased expression of immune-response genes (S100A9, DEFB4A, IL-4R) following microorganism exposure; however, there were marked differences between the unpigmented and pigmented skin equivalents. This physiologically relevant human 3D-skin equivalent opens up new avenues for studying complex skin pigmentation disorders, melanoma, and UV damage, as well as the rapidly evolving field of the skin microbiome and the balance between commensal and pathogenic species.
Collapse
Affiliation(s)
- Omera Bi
- Centre for Skin Sciences, Faculty of Life ScienceUniversity of BradfordBradfordUK
- Labskin UK, York Biotech CampusSand HuttonUK
| | | | - Stephen Sikkink
- Centre for Skin Sciences, Faculty of Life ScienceUniversity of BradfordBradfordUK
| | - Gill Westgate
- Centre for Skin Sciences, Faculty of Life ScienceUniversity of BradfordBradfordUK
| | - Sobia Kauser
- Centre for Skin Sciences, Faculty of Life ScienceUniversity of BradfordBradfordUK
| | - Jacobo Elies
- Faculty of Life SciencesUniversity of BradfordBradfordUK
| | - M. Julie Thornton
- Centre for Skin Sciences, Faculty of Life ScienceUniversity of BradfordBradfordUK
| |
Collapse
|
15
|
Liu K, Deng S, Zhou Y, Xu B, Zhang Y, Li W, Liu X, Yao X. Crosstalk Between the Skin Environment and Microbial Community in Immune-Related Skin Diseases. Clin Rev Allergy Immunol 2025; 68:16. [PMID: 39954089 DOI: 10.1007/s12016-025-09029-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/04/2025] [Indexed: 02/17/2025]
Abstract
The skin surface hosts diverse skin microbiota, including bacteria, fungi, and viruses. Intricate interactions between the skin microenvironment and microbial community are crucial for maintaining cutaneous homeostasis. This review explores the bidirectional relationship between the skin ecosystem and its microbiota. The skin microenvironment is shaped by a combination of intrinsic factors, dominated by sweat glands and pilosebaceous units, and external factors, such as UV radiation and personal care products, which create distinct niches that influence microbial colonization patterns across different skin regions. The skin microbiome, in turn, modulates the physical, chemical, immunological, and microbial barriers of the skin. We also discuss the alterations in this crosstalk in various immune-related skin conditions such as atopic dermatitis, psoriasis, rosacea, hidradenitis suppurativa, skin cancer, and aging. Understanding these interactions is vital for developing targeted microbiome-based therapies for various skin disorders. Further researches are needed to deepen insights into the microbial roles and their therapeutic potentials in skin health and disease.
Collapse
Affiliation(s)
- Kecheng Liu
- Department of Allergy and Rheumatology, Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, 210042, China
| | - Shuting Deng
- Department of Allergy and Rheumatology, Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, 210042, China
| | - Yuan Zhou
- Department of Allergy and Rheumatology, Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, 210042, China
| | - Beilei Xu
- Department of Allergy and Rheumatology, Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, 210042, China
| | - Yu Zhang
- Department of Allergy and Rheumatology, Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, 210042, China
| | - Wei Li
- Department of Dermatology, Institute of Dermatology, Huashan Hospital, Fudan University, Shanghai, Shanghai, 200040, China
| | - Xiaochun Liu
- Department of Allergy and Rheumatology, Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, 210042, China.
| | - Xu Yao
- Department of Allergy and Rheumatology, Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Hospital for Skin Diseases, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, 210042, China.
| |
Collapse
|
16
|
Alperovitz C, Ben David N, Ramot Y, Gross A, Mizrahi B. Living Microneedles for Intradermal Delivery of Beneficial Bacteria. ACS Biomater Sci Eng 2025; 11:1232-1241. [PMID: 39828947 PMCID: PMC11815633 DOI: 10.1021/acsbiomaterials.4c02230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2024] [Revised: 01/05/2025] [Accepted: 01/10/2025] [Indexed: 01/22/2025]
Abstract
The skin, our first line of defense against external threats, combines a physical barrier and a rich microbial community. Disruptions of this community, for example, due to infectious injury, have been linked to a decrease in bacteria diversity and to mild to severe pathological conditions. Although some progress has been made in the field, possibilities/procedures for restoring the skin microbiome are still far from ideal. The objective of this study was to design and evaluate a dissolvable poly(vinyl alcohol)/polyvinylpyrrolidone microneedle (MN) patch containing live Bacillus subtilis. According to the plan, bacteria were distributed equally throughout the patch without compromising the morphology and mechanical properties of the needles. B. subtilis was successfully released from the MNs, reaching a logarithmic growth phase after 5 h. These MNs demonstrated remarkable antibacterial activity against the Gram-positive pathogenic S. pyogenes, S. aureus, and C. acnes, while the empty control MNs showed no such activity. Finally, mice were inserted with a single MN patch loaded with GFP-B. subtilis presented significantly higher total radiance efficiency (TRE) values compared to the empty-MN mice throughout the entire experiment. This concept of incorporating live, secreting bacteria within a supportive MN patch shows great promise as a bacterial delivery system, offering a potential shift from conventional pharmacological approaches to more sustainable and symbiotic therapies.
Collapse
Affiliation(s)
- Caroline
Hali Alperovitz
- Faculty
of Biotechnology and Food Engineering, Technion—Israel
Institute of Technology, Haifa 3200003, Israel
| | - Noa Ben David
- Faculty
of Biotechnology and Food Engineering, Technion—Israel
Institute of Technology, Haifa 3200003, Israel
| | - Yuval Ramot
- Department
of Dermatology and Venereology, Hadassah−Hebrew
University Medical Center, Jerusalem 9112001, Israel
| | - Adi Gross
- Faculty
of Biotechnology and Food Engineering, Technion—Israel
Institute of Technology, Haifa 3200003, Israel
| | - Boaz Mizrahi
- Faculty
of Biotechnology and Food Engineering, Technion—Israel
Institute of Technology, Haifa 3200003, Israel
| |
Collapse
|
17
|
Scharschmidt TC, Segre JA. Skin microbiome and dermatologic disorders. J Clin Invest 2025; 135:e184315. [PMID: 39895627 PMCID: PMC11785926 DOI: 10.1172/jci184315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2025] Open
Abstract
Human skin acts as a physical barrier to prevent the entry of pathogenic microbes while simultaneously providing a home for commensal bacteria and fungi. Microbiome sequencing studies have demonstrated the unappreciated diversity and selectivity of these microbes. Functional studies have demonstrated the impact of specific strains to tune the immune system, sculpt the microbial community, provide colonization resistance, and promote epidermal barrier integrity. Recent studies have integrated the microbiome, immunity, and tissue integrity to understand their interplay in common disorders such as atopic dermatitis. In this Review, we explore microbiome shifts associated with cutaneous disorders with an eye toward how the microbiome can be mined to identify new therapeutic opportunities.
Collapse
Affiliation(s)
- Tiffany C. Scharschmidt
- Department of Dermatology, University of California, San Francisco, San Francisco, California, USA
| | - Julia A. Segre
- Microbial Genomics Section, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
18
|
Faccin M, O'Neill AM, Lawhon SD, Worthing KA, Wiener DJ, Gallo RL, Hoffmann AR. Staphylococcus felis C4 exhibits in vitro antimicrobial activity against methicillin-resistant Staphylococcus pseudintermedius in a novel canine skin explant model. Vet Dermatol 2025; 36:24-33. [PMID: 39450713 DOI: 10.1111/vde.13308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 07/19/2024] [Accepted: 10/08/2024] [Indexed: 10/26/2024]
Abstract
BACKGROUND Canine superficial pyoderma is a common bacterial skin infection of dogs, generally caused by Staphylococcus pseudintermedius. The C4 strain of Staphylococcus felis was recently discovered to have strong antimicrobial activity against S. pseudintermedius in mice. OBJECTIVES We aimed to evaluate in vitro if this antimicrobial activity was maintained using a novel canine skin explant model. MATERIALS AND METHODS Punch biopsies (8 mm) of skin from recently euthanised dogs were collected and placed into six-well plates on top of an agarose pedestal. RESULTS Histological examination of the skin explants showed an intact dermal-epidermal organisation and a stratum corneum that was successfully colonised by S. pseudintermedius after topical application. The number of colony forming units of S. pseudintermedius showed a 2 log increase after 24 h colonisation, indicating that the explant supported bacterial growth. By contrast, co-treatment with S. felis C4 live bacteria and its sterile protein product significantly reduced the growth of a methicillin-susceptible (ST540, p = 0.0357) and a methicillin-resistant (MR) strain (ST71, p = 0.0143) of S. pseudintermedius. No detectable bacteria were recovered from or visualised on skin 24 h posttreatment with the S. felis C4 sterile protein product. CONCLUSIONS AND CLINICAL RELEVANCE Using a novel canine explant model, we demonstrate that the S. felis C4 strain inhibits the growth of S. pseudintermedius and that it is a promising candidate for a new probiotic therapy to treat cutaneous infections caused by S. pseudintermedius, including MR strains.
Collapse
Affiliation(s)
- Mayane Faccin
- Department of Veterinary Pathobiology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, Texas, USA
| | - Alan M O'Neill
- Evotec (UK) Ltd., In Vitro Biology, Abingdon, Oxfordshire, UK
| | - Sara D Lawhon
- Department of Veterinary Pathobiology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, Texas, USA
| | - Kate A Worthing
- Sydney School of Veterinary Science, Faculty of Science, The University of Sydney, Camperdown, New South Wales, Australia
- The University of Sydney Institute of Infectious Diseases, Sydney, New South Wales, Australia
| | - Dominique J Wiener
- Department of Veterinary Pathobiology, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, Texas, USA
| | - Richard L Gallo
- Department of Dermatology, University of California-San Diego, San Diego, California, USA
| | - Aline Rodrigues Hoffmann
- Department of Comparative, Diagnostic, and Population Medicine, College of Veterinary Medicine, University of Florida, Gainesville, Florida, USA
| |
Collapse
|
19
|
Selvaraj S, Perera M, Yapa P, Munaweera I, Perera IC, Senapathi T, Weerasinghe L. In vitro analysis of XLAsp-P2 peptide loaded cellulose acetate nanofiber for wound healing. J Pharm Sci 2025; 114:911-922. [PMID: 39542360 DOI: 10.1016/j.xphs.2024.10.050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 10/28/2024] [Accepted: 10/28/2024] [Indexed: 11/17/2024]
Abstract
Recently, nanofiber-based wound dressings are currently a viable strategy to expedite the healing of wounds by providing a suitable microenvironment for tissue growth with active ingredients. This research study subjects the development of electrospun cellulose acetate (CA) nanofibers loaded with the XLAsp-P2, an antimicrobial peptide (AMP) that holds great potential for enhanced wound healing as a therapeutic agent. The synthesized XLAsp-P2-loaded CA nanofibers were fabricated via three loading percentages, 0.1 %, 0.2 %, and 0.3 % w/w, and characterized and evaluated their antimicrobial potential with MTT assay and Agar overlay methods as an alternative strategy. FT-IR analysis confirmed the compatibility of the peptide-loaded CA nanocomposite, showing distinct peaks corresponding to the constituent materials. Scanning electron microscopy (SEM) analysis was employed to characterize the morphology of electrospun peptide CA nanocomposites and illustrate the fiber's size at the nanoscale. The in vitro release study during the 24 hr, 87 % of the peptide was released which was approximately 5.2 mg; which was closer matched to the square root model of Higuchi at room temperature. MTT assay presented sensitive results towards Gram-positive bacteria compared to Gram Negative bacteria; which corresponded to the inhibition zones of the Agar overlay method proving that Escherichia coli (ATCC 25922) 17.66 ± 0.38 mm and Pseudomonas aeruginosa (ATCC 27853) 17.44 ± 0.38 mm exhibited moderate susceptibility, while Staphylococcus aureus (ATCC 25923)19.89 ± 0.69 mm and Bacillus cereus (ATCC 11778) 23.00 ± 0.33 mm showed promising responses. Collectively, The study's findings indicate that the XLAsp-P2 incorporated CA mat possesses an opportunity to function as an efficient platform for delivering therapeutic peptides.
Collapse
Affiliation(s)
- Saranya Selvaraj
- Department of Chemistry, Faculty of Applied Sciences, University of Sri Jayewardenepura, Gangodawila, Nugegoda, Sri Lanka
| | - Monali Perera
- Department of Chemistry, Faculty of Applied Sciences, University of Sri Jayewardenepura, Gangodawila, Nugegoda, Sri Lanka
| | - Piumika Yapa
- Department of Chemistry, Faculty of Applied Sciences, University of Sri Jayewardenepura, Gangodawila, Nugegoda, Sri Lanka
| | - Imalka Munaweera
- Department of Chemistry, Faculty of Applied Sciences, University of Sri Jayewardenepura, Gangodawila, Nugegoda, Sri Lanka
| | - Inoka C Perera
- Department of Zoology and Environment Sciences, Faculty of Science, University of Colombo, Colombo, Sri Lanka
| | - Tharindu Senapathi
- Department of Chemistry, Faculty of Applied Sciences, University of Sri Jayewardenepura, Gangodawila, Nugegoda, Sri Lanka
| | - Laksiri Weerasinghe
- Department of Chemistry, Faculty of Applied Sciences, University of Sri Jayewardenepura, Gangodawila, Nugegoda, Sri Lanka.
| |
Collapse
|
20
|
Shao L, Li T, Yang S, Ma L, Cai B, Jia Q, Jiang H, Bai T, Li Y. The prebiotic effects of fructooligosaccharides enhance the growth characteristics of Staphylococcus epidermidis and enhance the inhibition of Staphylococcus aureus biofilm formation. Int J Cosmet Sci 2025; 47:155-167. [PMID: 39246292 DOI: 10.1111/ics.13020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2024] [Revised: 08/07/2024] [Accepted: 08/19/2024] [Indexed: 09/10/2024]
Abstract
OBJECTIVE Oligosaccharides have been shown to enhance the production of short chain fatty acids (SCFAs) by gut probiotics and regulate gut microbiota, to improve intestinal health. Recent research indicates that oligosaccharides may also positively impact skin microbiota by selectively promoting the growth of skin commensal bacteria and inhibiting pathogenic bacteria. However, the specific metabolic and regulatory mechanisms of skin commensal bacteria in response to oligosaccharides remain unclear. This study aims to explore the influence of four oligosaccharides on the growth and metabolism of Staphylococcus epidermidis and further identify skin prebiotics that can enhance its probiotic effects on the skin. METHODS Fructooligosaccharides (FOS), isomaltooligosaccharide (IMO), galactooligosaccharides (GOS) and inulin were compared in terms of their impact on cell proliferation, SCFAs production of S. epidermidis CCSM0287 and the biofilm inhibition effect of their fermentation supernatants on Staphylococcus aureus CCSM0424. Furthermore, the effect of FOS on S. epidermidis CCSM0287 was analysed by the transcriptome analysis. RESULTS All four oligosaccharides effectively promoted the growth of S. epidermidis CCSM0287 cells, increased the production of SCFAs, with FOS demonstrating the most significant effect. Analysis of the SCFAs indicated that S. epidermidis CCSM0287 predominantly employs oligosaccharides to produce acetic acid and isovaleric acid, differing from the SCFAs produced by gut microbiota. Among the four oligosaccharides, the addition of 2% FOS fermentation supernatant significantly inhibited S. aureus CCSM0424 biofilm formation. Furthermore, RNA sequencing revealed 162 differentially expressed genes (84 upregulated and 78 downregulated) of S. epidermidis CCSM0287 upon FOS treatment compared with glucose treatment. Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis highlighted differences in the amino acid synthesis pathway, particularly in terms of arginine biosynthesis. CONCLUSION FOS promotes cell proliferation, increases the SCFA production of S. epidermidis CCSM0287 and enhance the inhibition of S. aureus biofilm formation, suggesting that FOS serves as a potential prebiotic for strain S. epidermidis CCSM0287.
Collapse
Affiliation(s)
- Li Shao
- School of Perfume and Aroma Technology, Shanghai Institute of Technology, Shanghai, China
| | - Tao Li
- School of Perfume and Aroma Technology, Shanghai Institute of Technology, Shanghai, China
| | - Suzhen Yang
- R&D Innovation Center, Shandong Freda Biotech Co., Ltd., Jinan, Shandong, China
| | - Laiji Ma
- School of Perfume and Aroma Technology, Shanghai Institute of Technology, Shanghai, China
| | - Banruo Cai
- School of Perfume and Aroma Technology, Shanghai Institute of Technology, Shanghai, China
| | - Qingwen Jia
- R&D Innovation Center, Shandong Freda Biotech Co., Ltd., Jinan, Shandong, China
| | - Hong Jiang
- R&D Innovation Center, Shandong Freda Biotech Co., Ltd., Jinan, Shandong, China
| | - Tianming Bai
- R&D Innovation Center, Shandong Freda Biotech Co., Ltd., Jinan, Shandong, China
| | - Yan Li
- R&D Innovation Center, Shandong Freda Biotech Co., Ltd., Jinan, Shandong, China
| |
Collapse
|
21
|
Uberoi A, Murga-Garrido SM, Bhanap P, Campbell AE, Knight SAB, Wei M, Chan A, Senay T, Tegegne S, White EK, Sutter CH, Mesaros C, Sutter TR, Grice EA. Commensal-derived tryptophan metabolites fortify the skin barrier: Insights from a 50-species gnotobiotic model of human skin microbiome. Cell Chem Biol 2025; 32:111-125.e6. [PMID: 39824155 PMCID: PMC11753614 DOI: 10.1016/j.chembiol.2024.12.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Revised: 11/01/2024] [Accepted: 12/12/2024] [Indexed: 01/20/2025]
Abstract
The epidermal barrier defends the body against dehydration and harmful substances. The commensal microbiota is essential for proper differentiation and repair of the epidermal barrier, an effect mediated by the aryl hydrocarbon receptor (AHR). However, the microbial mechanisms of AHR activation in skin are less understood. Tryptophan metabolites are AHR ligands that can be products of microbial metabolism. To identify microbially regulated tryptophan metabolites in vivo, we established a gnotobiotic model colonized with fifty human skin commensals and performed targeted mass spectrometry on murine skin. Indole-related metabolites were enriched in colonized skin compared to germ-free skin. In reconstructed human epidermis and in murine models of atopic-like barrier damage, these metabolites improved barrier repair and function individually and as a cocktail. These results provide a framework for the identification of microbial metabolites that mediate specific host functions, which can guide the development of microbe-based therapies for skin disorders.
Collapse
Affiliation(s)
- Aayushi Uberoi
- Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Department of Pathology and Immunology, Division of Laboratory and Genomic Medicine, Washington University School of Medicine in St. Louis, St. Louis, MO, USA.
| | - Sofía M Murga-Garrido
- Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Preeti Bhanap
- Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Amy E Campbell
- Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Simon A B Knight
- Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Monica Wei
- Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Anya Chan
- Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Taylor Senay
- Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Saba Tegegne
- Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ellen K White
- Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | | | - Clementina Mesaros
- Center of Excellence in Environmental Toxicology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Thomas R Sutter
- Department of Biological Sciences, University of Memphis, Memphis, TN, USA
| | - Elizabeth A Grice
- Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
22
|
Huang C, Zhuo F, Guo Y, Wang S, Zhang K, Li X, Dai W, Dou X, Yu B. Skin microbiota: pathogenic roles and implications in atopic dermatitis. Front Cell Infect Microbiol 2025; 14:1518811. [PMID: 39877655 PMCID: PMC11772334 DOI: 10.3389/fcimb.2024.1518811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Accepted: 12/27/2024] [Indexed: 01/31/2025] Open
Abstract
Atopic dermatitis (AD) is a chronic and inflammatory skin disorder characterized by impaired barrier function and imbalanced immunity. Recent advances have revealed that dysbiosis of skin microbiota plays important roles in the pathogenesis and development of AD. Meanwhile, endogenous and external factors contribute to the dysbiosis of skin microbiota in AD. Additionally, various treatments, including topical treatments, phototherapy, and systemic biologics, have demonstrated positive impacts on the clinical outcomes, alongside with the modulations of cutaneous microbiota in AD patients. Importantly, therapeutics or products regulating skin microbiota homeostasis have demonstrated potential for AD treatment in early clinical studies. In this review, we underline changes of the skin microbiota correlated with AD. Meanwhile, we provide an overview of the skin microbiota regarding its roles in the pathogenesis and development of AD. Finally, we summarize therapeutic strategies restoring the skin microbial homeostasis in AD management.
Collapse
Affiliation(s)
- Cong Huang
- Department of Dermatology, Skin Research Institute of Peking University Shenzhen Hospital, Peking University Shenzhen Hospital, Shenzhen, China
- Shenzhen Key Laboratory for Translational Medicine of Dermatology, Shenzhen Peking University - the Hong Kong University of Science and Technology Medical Center, Shenzhen, China
| | - Fan Zhuo
- Department of Dermatology, Skin Research Institute of Peking University Shenzhen Hospital, Peking University Shenzhen Hospital, Shenzhen, China
- Shenzhen Key Laboratory for Translational Medicine of Dermatology, Shenzhen Peking University - the Hong Kong University of Science and Technology Medical Center, Shenzhen, China
| | - Yang Guo
- Department of Dermatology, Skin Research Institute of Peking University Shenzhen Hospital, Peking University Shenzhen Hospital, Shenzhen, China
- Shenzhen Key Laboratory for Translational Medicine of Dermatology, Shenzhen Peking University - the Hong Kong University of Science and Technology Medical Center, Shenzhen, China
- Department of Epidemiology and Statistics, School of Public Health, Hebei Key Laboratory of Environment and Human Health, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Siyu Wang
- Department of Dermatology, Skin Research Institute of Peking University Shenzhen Hospital, Peking University Shenzhen Hospital, Shenzhen, China
- Shenzhen Key Laboratory for Translational Medicine of Dermatology, Shenzhen Peking University - the Hong Kong University of Science and Technology Medical Center, Shenzhen, China
- Department of Dermatology, Peking University Shenzhen Hospital, Shenzhen, China
- Department of Dermatology, Peking University Shenzhen Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| | - Kaoyuan Zhang
- Department of Dermatology, Skin Research Institute of Peking University Shenzhen Hospital, Peking University Shenzhen Hospital, Shenzhen, China
- Shenzhen Key Laboratory for Translational Medicine of Dermatology, Shenzhen Peking University - the Hong Kong University of Science and Technology Medical Center, Shenzhen, China
| | - Xiahong Li
- Department of Dermatology, Skin Research Institute of Peking University Shenzhen Hospital, Peking University Shenzhen Hospital, Shenzhen, China
- Shenzhen Key Laboratory for Translational Medicine of Dermatology, Shenzhen Peking University - the Hong Kong University of Science and Technology Medical Center, Shenzhen, China
| | - Wenkui Dai
- Department of Obstetrics and Gynecology, Peking University Shenzhen Hospital, Shenzhen, China
| | - Xia Dou
- Department of Dermatology, Skin Research Institute of Peking University Shenzhen Hospital, Peking University Shenzhen Hospital, Shenzhen, China
- Shenzhen Key Laboratory for Translational Medicine of Dermatology, Shenzhen Peking University - the Hong Kong University of Science and Technology Medical Center, Shenzhen, China
| | - Bo Yu
- Department of Dermatology, Skin Research Institute of Peking University Shenzhen Hospital, Peking University Shenzhen Hospital, Shenzhen, China
- Shenzhen Key Laboratory for Translational Medicine of Dermatology, Shenzhen Peking University - the Hong Kong University of Science and Technology Medical Center, Shenzhen, China
| |
Collapse
|
23
|
Huang RY, Zhang C, Lim HL. Ribotyping Staphylococcus epidermidis Using Probabilistic Sequence Analysis and Levenshtein Distance Algorithm. Curr Microbiol 2025; 82:78. [PMID: 39792222 PMCID: PMC11723854 DOI: 10.1007/s00284-024-04057-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Accepted: 12/25/2024] [Indexed: 01/12/2025]
Abstract
Staphylococcus epidermidis (S. epidermidis) live in different human locations and natural environments. For ribotyping S. epidermidis sub-species, 2507 PCR-amplified reads of 16S rRNA genes of S. epidermidis in a public dataset were used for probabilistic sequence analysis. A sequence probability logo (sequence pLogo) as a reference sequence of 16S rRNA genes of S. epidermidis was constructed. Through implementation of Levenshtein Distance algorithm, two 20-base pairs (bp) motifs, commonly present in 2507 PCR-amplified reads, were identified. The top 38 S. epidermidis isolates, which carried 16S rRNA nucleotide domains that were made of different sequences but have high similarity scores to two 20-bp motifs, were found from 11 human, 8 animal, 9 plant and 10 environmental samples, indicating that these two 20-bp motifs were broadly present in diverse S. epidermidis isolates. Thirty-one PCR-amplified reads of 16S rRNA genes, which were currently not in the dataset, were utilized to verify the feasibility of using two 20-bp motifs for ribotyping S. epidermidis sub-species. S. epidermidis S1, S3, but not S2, isolates on the human scalp carried a 20-bp sequence domain with high similarities to a 20-bp motif in the sequence pLogo. The phylogenetic tree showed that S. epidermidis S1, S2 and S3 were not from a single common ancestor. Two newly identified 20-bp motifs here, thus, provided reference nucleotide residues for ribotyping S. epidermidis.
Collapse
Affiliation(s)
- Ryan Yuki Huang
- Department of Computer Science, Program in Liberal Medical Education (PLME), Brown University, Providence, RI, 02906, USA.
| | - Chengye Zhang
- Information Science and Technology, Universiti Kebangsaan, 43600, Lingkungan Ilmu, Selangor, Malaysia
| | | |
Collapse
|
24
|
Dafinone ME, Lyle RE, Lee C, Mehta A, Dahle SE, Isseroff RR. Non-antibiotic approaches to mitigating wound infections: Potential for SSRIs and adrenergic antagonists as emerging therapeutics. Wound Repair Regen 2025; 33:e13240. [PMID: 39737521 DOI: 10.1111/wrr.13240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 10/16/2024] [Accepted: 11/15/2024] [Indexed: 01/01/2025]
Abstract
Bacterial biofilms represent a formidable challenge in the treatment of chronic wounds, largely because of their resistance to conventional antibiotics. The emergence of multidrug-resistant (MDR) bacterial strains exacerbates this issue, necessitating a shift towards exploring alternative therapeutic approaches. In response to this urgent need, there has been a surge in research efforts aimed at identifying effective non-antibiotic treatments. Recently noted among the non-antibiotic options are selective serotonin reuptake inhibitors (SSRIs) and beta-adrenergic (β-AR) antagonists. Both have demonstrated antimicrobial activities and wound-healing properties, which makes them particularly promising potential therapeutics for chronic wounds. This review seeks to comprehensively evaluate the landscape of non-antibiotic strategies for managing wound infections. By analysing the latest research findings and clinical developments, it aims to shed light on emerging therapeutic alternatives. Additionally, the review delves into the potential of repurposing systemic therapeutics for topical application, offering insights into the feasibility and challenges associated with current approaches. We also address the necessity of translating promising preclinical results into tangible clinical benefits.
Collapse
Affiliation(s)
- Mirabel E Dafinone
- Department of Dermatology, University of California Davis School of Medicine, Sacramento, California, USA
- School of Medicine, University of Nevada Reno, Reno, Nevada, USA
- Dermatology Section, VA Northern California Health Care System, McClellan Park, California, USA
| | - Rawlings E Lyle
- Department of Dermatology, University of California Davis School of Medicine, Sacramento, California, USA
- Dermatology Section, VA Northern California Health Care System, McClellan Park, California, USA
- School of Medicine, University of California Davis, Davis, California, USA
| | - Conan Lee
- School of Medicine, University of California Davis, Davis, California, USA
| | - Alisha Mehta
- Dermatology Section, VA Northern California Health Care System, McClellan Park, California, USA
- College of Medicine, California Northstate University, Elk Grove, California, USA
| | - Sara E Dahle
- School of Medicine, University of California Davis, Davis, California, USA
- Podiatry Section, VA Northern California Health Care System, McClellan Park, California, USA
| | - R Rivkah Isseroff
- Department of Dermatology, University of California Davis School of Medicine, Sacramento, California, USA
- Dermatology Section, VA Northern California Health Care System, McClellan Park, California, USA
| |
Collapse
|
25
|
Lee MT, Tan X, Le HH, Besler K, Thompson S, Harris-Tryon T, Johnson EL. Gut bacterial sphingolipid production modulates dysregulated skin lipid homeostasis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.29.629238. [PMID: 39803564 PMCID: PMC11722302 DOI: 10.1101/2024.12.29.629238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/16/2025]
Abstract
Sphingolipids are an essential lipid component of the skin barrier with alterations in skin sphingolipid composition associated with multiple skin disorders including psoriasis, atopic dermatitis, and ichthyosis. Contributions to skin sphingolipid abundance are not well characterized, thus the main method of modulating skin lipid levels is the topical application of creams rich with sphingolipids at the skin surface. Evidence that diet and gut microbiome function can alter skin biology proposes an intriguing potential for the modulation of skin lipid homeostasis through gut microbial metabolism, but potential mechanisms of action are not well understood. Sphingolipid synthesis by prominent gut microbes has been shown to affect intestinal, hepatic and immune functions with the potential for sphingolipid-producing bacteria to affect skin biology through altering skin sphingolipid levels. To address this question, we used bioorthogonal chemistry to label lipids from the sphingolipid-producing bacteria Bacteroides thetaiotaomicron and trace these lipids to the skin epidermis. Exposing mice to B. thetaiotaomicron strains mutant in the ability to produce sphingolipids resulted in significantly lower transfer of gut microbiome-derived lipids to the skin, while also altering skin biology and altering expression of skin barrier genes. Measurement of skin ceramide levels, a class of sphingolipids involved in skin barrier function, determined that skin sphingolipid levels were altered in the presence of gut sphingolipid-producing bacteria. Together this work demonstrates that gut bacterial lipids can transfer to the skin and provides a compelling avenue for modulating sphingolipid-dominant compartments of the skin using sphingolipid-producing bacteria of the gut microbiome.
Collapse
Affiliation(s)
- Min-Ting Lee
- Divison of Nutritional Sciences, Cornell University, Ithaca, NY, USA
| | - Xiaoqing Tan
- Divison of Nutritional Sciences, Cornell University, Ithaca, NY, USA
| | - Henry H. Le
- Divison of Nutritional Sciences, Cornell University, Ithaca, NY, USA
| | - Kevin Besler
- Divison of Nutritional Sciences, Cornell University, Ithaca, NY, USA
- Howard Hughes Medical Institute, Cornell University, Ithaca, NY, USA
| | - Sharon Thompson
- Divison of Nutritional Sciences, Cornell University, Ithaca, NY, USA
- Howard Hughes Medical Institute, Cornell University, Ithaca, NY, USA
| | - Tamia Harris-Tryon
- Department of Dermatology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Elizabeth L. Johnson
- Divison of Nutritional Sciences, Cornell University, Ithaca, NY, USA
- Howard Hughes Medical Institute, Cornell University, Ithaca, NY, USA
| |
Collapse
|
26
|
Zhang Z, Wu W, Lin J, Li H. Unveiling the hidden causal links: skin flora and cutaneous melanoma. Front Oncol 2024; 14:1451175. [PMID: 39723372 PMCID: PMC11668787 DOI: 10.3389/fonc.2024.1451175] [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: 06/18/2024] [Accepted: 11/25/2024] [Indexed: 12/28/2024] Open
Abstract
Objective The presence of skin flora (SF) has been identified as a significant factor in the onset and progression of cutaneous melanoma (CM). However, the vast diversity and abundance of SF present challenges to fully understanding the causal relationship between SF and CM. Methods A Two Sample Mendelian Randomization (TSMR) analysis was conducted to investigating the causal relationship between SF and CM. The Inverse-Variance Weighted (IVW) method was utilized as the primary approach to assess the causal relationship under investigation. Furthermore, an independent external cohort was employed to validate the initial findings, followed by a meta-analysis of the consolidated results. To address potential confounding factors related to the influence of SF on CM, a Multivariate Mendelian Randomization (MVMR) analysis was also conducted. Finally, a Reverse Mendelian Randomization (RMR) was conducted to further validate the causal association. Results TSMR results showed that 9 SF have a causal relationship with CM in the training cohort. Although these 9 SF weren't confirmed in the testing cohort, 4 SF remained significant in the meta-analysis after integrating results from both cohorts. MVMR analysis indicated that 3 SF were still significantly associated with CM after accounting for the interactions between different SF in the training cohort. No reverse causal relationship was identified in RMR analysis. Conclusion A total of 9 SF were identified as having a potential causal relationship with CM; however, a large randomized controlled trial is needed to verify these results.
Collapse
Affiliation(s)
- Zexin Zhang
- The Second Clinical School of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Wenfeng Wu
- The Second Clinical School of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jiajia Lin
- The Second Clinical School of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Hongyi Li
- Department of Dermatology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
27
|
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
|
28
|
Bay L, Jemec GB, Ring HC. Microenvironmental host-microbe interactions in chronic inflammatory skin diseases. APMIS 2024; 132:974-984. [PMID: 39270740 PMCID: PMC11582343 DOI: 10.1111/apm.13464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Accepted: 08/12/2024] [Indexed: 09/15/2024]
Abstract
Several microbiome studies have recently demonstrated microbial dysbiosis in various chronic inflammatory skin diseases, and it is considered an important role in the pathogenesis. Although the role of skin dysbiosis in inflammatory skin diseases is debatable, the local microenvironment is considered essential concerning compositional changes and functional alterations of the skin microbiota. Indeed, various local nutrients (e.g., lipids), pH values, water, oxygen, and antimicrobial peptides may affect the level of skin dysbiosis in these skin diseases. In particular, in atopic dermatitis and hidradenitis suppurativa, significant changes in skin dysbiosis have been associated with local aberrant host immune changes. In this review, the potential pathogenic crosstalk between the host and the microbiota is reviewed in relation to the physical, chemical, and biological microenvironments of various chronic inflammatory skin diseases.
Collapse
Affiliation(s)
- Lene Bay
- Bacterial Infection Biology, Department of Immunology and Microbiology, Costerton Biofilm Center, Faculty of Health and Medical SciencesUniversity of CopenhagenCopenhagenDenmark
| | - Gregor Borut Jemec
- Department of DermatologyZealand University HospitalRoskildeDenmark
- Faculty of Health and Medical SciencesUniversity of CopenhagenCopenhagenDenmark
| | | |
Collapse
|
29
|
Sasikumar J, P P K, Naik B, Das SP. A greener side of health care: Revisiting phytomedicine against the human fungal pathogen Malassezia. Fitoterapia 2024; 179:106243. [PMID: 39389474 DOI: 10.1016/j.fitote.2024.106243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 10/02/2024] [Accepted: 10/05/2024] [Indexed: 10/12/2024]
Abstract
Malassezia species are commensal fungi residing on the skin and in the gut of humans and animals. Yet, under certain conditions, they become opportunistic pathogens leading to various clinical conditions including dermatological disorders. The emergence of drug resistance and adverse effects associated with conventional antifungal agents has propelled the search for alternative treatments, among which phytomedicine stands out prominently. Phytochemicals, including phenolic acids, flavonoids, and terpenoids, demonstrate potential antifungal activity against Malassezia by inhibiting its growth, adhesion, and biofilm formation. Furthermore, the multifaceted therapeutic properties of phytomedicine (including anti-fungal and, antioxidant properties) contribute to its efficacy in alleviating symptoms associated with Malassezia infections. Despite these promising prospects, several challenges hinder the widespread adoption of phytomedicine in clinical practice mostly since the mechanistic studies and controlled experiments to prove efficacy have not been done. Issues include standardization of herbal extracts, variable bioavailability, and limited clinical evidence. Hence, proper regulatory constraints necessitate comprehensive research endeavors and regulatory frameworks to harness the full therapeutic potential of phytomedicine. In conclusion, while phytomedicine holds immense promise as an alternative or adjunctive therapy against Malassezia, addressing these challenges is imperative to optimize its efficacy and ensure its integration into mainstream medical care. In this review we provide an update on the potential phytomedicines in combating Malassezia-related ailments, emphasizing its diverse chemical constituents and mechanisms of action.
Collapse
Affiliation(s)
- Jayaprakash Sasikumar
- Cell Biology and Molecular Genetics, Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore 575018, India
| | - Keerthana P P
- Cell Biology and Molecular Genetics, Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore 575018, India
| | - Bharati Naik
- Cell Biology and Molecular Genetics, Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore 575018, India
| | - Shankar Prasad Das
- Cell Biology and Molecular Genetics, Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore 575018, India.
| |
Collapse
|
30
|
Ferrara F, Valacchi G. Role of microbiota in the GUT-SKIN AXIS responses to outdoor stressors. Free Radic Biol Med 2024; 225:894-909. [PMID: 39505118 DOI: 10.1016/j.freeradbiomed.2024.11.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 10/31/2024] [Accepted: 11/02/2024] [Indexed: 11/08/2024]
Abstract
Beside the respiratory tract, the skin and the gut represent the first defensive lines of our body against the external insults displaying many important biochemical features able to maintain the epithelial barrier integrity and to regulate the tissue immune responses. The human microbiome is essential in maintaining the tissue homeostasis and its dysregulation may lead to tissue conditions including inflammatory pathologies. Among all external insults, air pollutants have been shown to cause oxidative stress damage within the target tissues via an OxInflammatory response. Dysregulation of the gut microbiome (dysbiosis) by outdoor stressors, including air pollutants, may promote the exacerbation of the skin tissue damage via the interplay between the gut-skin axis. The intent of this review is to highlight the ability of exogenous stressors to modulate the human gut-skin axis via a redox regulated mechanism affecting the microbiome and therefore contributing to the development and aggravation of gut and skin conditions.
Collapse
Affiliation(s)
- Francesca Ferrara
- Department of Chemical, Pharmaceuticals and Agricultural Sciences, University of Ferrara, 44121, Ferrara, Italy
| | - Giuseppe Valacchi
- Department of Environmental and Prevention Sciences, University of Ferrara, 44121, Ferrara, Italy; Department of Animal Sciences, Plants for Human Health Institute, NC Research Campus, NC State University, Kannapolis, NC, 28081, USA; Kyung Hee University, Department of Food and Nutrition, Seoul, South Korea.
| |
Collapse
|
31
|
Nguyen UT, Salamzade R, Sandstrom S, Swaney MH, Townsend L, Wu SY, Cheong JA, Sardina JA, Ludwikoski I, Rybolt M, Wan H, Carlson C, Zarnowski R, Andes D, Currie C, Kalan L. Large-scale investigation for antimicrobial activity reveals novel defensive species across the healthy skin microbiome. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.04.621544. [PMID: 39574598 PMCID: PMC11580923 DOI: 10.1101/2024.11.04.621544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/30/2024]
Abstract
The human skin microbiome constitutes a dynamic barrier that can impede pathogen invasion by producing antimicrobial natural products. Gene clusters encoding for production of secondary metabolites, biosynthetic gene clusters (BGCs), that are enriched in the human skin microbiome relative to other ecological settings, position this niche as a promising source for new natural product mining. Here, we introduce a new human microbiome isolate collection, the EPithelial Isolate Collection (EPIC). It includes a large phylogenetically diverse set of human skin-derived bacterial strains from eight body sites. This skin collection, consisting of 980 strains is larger and more diverse than existing resources, includes hundreds of rare and low-abundance strains, and hundreds of unique BGCs. Using a large-scale co-culture screen to assess 8,756 pairwise interactions between skin-associated bacteria and potential pathogens, we reveal broad antifungal activity by skin microbiome members. Integrating 287 whole isolate genomes and 268 metagenomes from sampling sites demonstrates that while the distribution of BGC types is stable across body sites, specific gene cluster families (GCFs), each predicted to encode for a distinct secondary metabolite, can substantially vary. Sites that are dry or rarely moist harbor the greatest potential for discovery of novel bioactive metabolites. Among our discoveries are four novel bacterial species, three of which exert significant and broad-spectrum antifungal activity. This comprehensive isolate collection advances our understanding of the skin microbiomes biosynthetic capabilities and pathogen-fighting mechanisms, opening new avenues towards antimicrobial drug discovery and microbiome engineering.
Collapse
Affiliation(s)
- Uyen Thy Nguyen
- Department of Medical Microbiology and Immunology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Microbiology Doctoral Training Program, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
- M. G. DeGroote Institute for Infectious Disease Research, University of Wisconsin-Madison, Madison, USA
- David Braley Centre for Antibiotic Discovery, University of Wisconsin-Madison, Madison, USA
| | - Rauf Salamzade
- Department of Medical Microbiology and Immunology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Microbiology Doctoral Training Program, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Shelby Sandstrom
- Department of Medical Microbiology and Immunology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Mary Hannah Swaney
- Department of Medical Microbiology and Immunology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Microbiology Doctoral Training Program, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Liz Townsend
- Department of Medical Microbiology and Immunology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Microbiology Doctoral Training Program, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Sherrie Y. Wu
- Department of Medical Microbiology and Immunology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - J.Z. Alex Cheong
- Department of Medical Microbiology and Immunology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Microbiology Doctoral Training Program, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Joseph A. Sardina
- Department of Medical Microbiology and Immunology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Department of Bacteriology, College of Agriculture and Life Science, University of Wisconsin-Madison, Madison, USA
| | - Isabelle Ludwikoski
- Department of Medical Microbiology and Immunology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Mackinnley Rybolt
- Department of Medical Microbiology and Immunology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Hanxiao Wan
- Department of Medical Microbiology and Immunology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Caitlin Carlson
- Department of Bacteriology, College of Agriculture and Life Science, University of Wisconsin-Madison, Madison, USA
| | - Robert Zarnowski
- Department of Medicine, Division of Infectious Disease, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - David Andes
- Department of Medical Microbiology and Immunology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Department of Medicine, Division of Infectious Disease, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Cameron Currie
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
- M. G. DeGroote Institute for Infectious Disease Research, University of Wisconsin-Madison, Madison, USA
- David Braley Centre for Antibiotic Discovery, University of Wisconsin-Madison, Madison, USA
- Department of Bacteriology, College of Agriculture and Life Science, University of Wisconsin-Madison, Madison, USA
| | - Lindsay Kalan
- Department of Medical Microbiology and Immunology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
- M. G. DeGroote Institute for Infectious Disease Research, University of Wisconsin-Madison, Madison, USA
- David Braley Centre for Antibiotic Discovery, University of Wisconsin-Madison, Madison, USA
- Department of Medicine, Division of Infectious Disease, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, Wisconsin, USA
| |
Collapse
|
32
|
Dinić M, Verpile R, Burgess JL, Ming J, Marjanovic J, Beliz CN, Plano L, Hower S, Thaller SR, Banerjee S, Lev‐Tov H, Tomic‐Canic M, Pastar I. Multi-drug resistant Staphylococcus epidermidis from chronic wounds impair healing in human wound model. Wound Repair Regen 2024; 32:799-810. [PMID: 39439244 PMCID: PMC11584363 DOI: 10.1111/wrr.13231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 09/13/2024] [Accepted: 10/08/2024] [Indexed: 10/25/2024]
Abstract
Venous leg ulcers (VLUs) represent one of the most prevalent types of chronic wounds characterised by perturbed microbiome and biofilm-forming bacteria. As one of the most abundant skin-commensal, Staphylococcus epidermidis is known as beneficial for the host, however, some strains can form biofilms and hinder wound healing. In this study, S. epidermidis distribution in VLUs and associated resistome were analysed in ulcer tissue from patients. Virulence of S. epidermidis isolates from VLUs were evaluated by whole genome sequencing, antimicrobial susceptibility testing, in vitro biofilm and binding assays, and assessment of biofilm-forming capability and pro-inflammatory potential using human ex vivo wound model. We demonstrated that S. epidermidis isolates from VLUs inhibit re-epithelialization through biofilm-dependent induction of IL-1β, IL-8, and IL-6 which was in accordance with impaired healing outcomes observed in patients. High extracellular matrix binding ability of VLU isolates was associated with antimicrobial resistance and expression levels of the embp and sdrG, responsible for bacterial binding to fibrinogen and fibrin, respectively. Finally, we showed that S. epidermidis from VLUs demonstrate pathogenic features with ability to impair healing which underscores the emergence of treatment-resistant virulent lineages in patients with chronic ulcers.
Collapse
Affiliation(s)
- Miroslav Dinić
- Wound Healing and Regenerative Medicine Research Program, Dr. Phillip Frost Department of Dermatology and Cutaneous SurgeryUniversity of Miami Miller School of MedicineMiamiFloridaUSA
- Group for Probiotics and Microbiota‐Host Interaction, Institute of Molecular Genetics and Genetic EngineeringUniversity of BelgradeBelgradeSerbia
| | - Rebecca Verpile
- Wound Healing and Regenerative Medicine Research Program, Dr. Phillip Frost Department of Dermatology and Cutaneous SurgeryUniversity of Miami Miller School of MedicineMiamiFloridaUSA
| | - Jamie L. Burgess
- Wound Healing and Regenerative Medicine Research Program, Dr. Phillip Frost Department of Dermatology and Cutaneous SurgeryUniversity of Miami Miller School of MedicineMiamiFloridaUSA
| | - Jingjing Ming
- Wound Healing and Regenerative Medicine Research Program, Dr. Phillip Frost Department of Dermatology and Cutaneous SurgeryUniversity of Miami Miller School of MedicineMiamiFloridaUSA
| | - Jelena Marjanovic
- Wound Healing and Regenerative Medicine Research Program, Dr. Phillip Frost Department of Dermatology and Cutaneous SurgeryUniversity of Miami Miller School of MedicineMiamiFloridaUSA
| | - Carmen Nicole Beliz
- Wound Healing and Regenerative Medicine Research Program, Dr. Phillip Frost Department of Dermatology and Cutaneous SurgeryUniversity of Miami Miller School of MedicineMiamiFloridaUSA
| | - Lisa Plano
- Department of Microbiology and ImmunologyUniversity of Miami Miller School of MedicineMiamiFloridaUSA
| | - Suzanne Hower
- Department of Microbiology and ImmunologyUniversity of Miami Miller School of MedicineMiamiFloridaUSA
| | - Seth R. Thaller
- Department of SurgeryUniversity of Miami Miller School of MedicineMiamiFloridaUSA
| | - Santanu Banerjee
- Department of SurgeryUniversity of Miami Miller School of MedicineMiamiFloridaUSA
| | - Hadar Lev‐Tov
- Wound Healing and Regenerative Medicine Research Program, Dr. Phillip Frost Department of Dermatology and Cutaneous SurgeryUniversity of Miami Miller School of MedicineMiamiFloridaUSA
| | - Marjana Tomic‐Canic
- Wound Healing and Regenerative Medicine Research Program, Dr. Phillip Frost Department of Dermatology and Cutaneous SurgeryUniversity of Miami Miller School of MedicineMiamiFloridaUSA
| | - Irena Pastar
- Wound Healing and Regenerative Medicine Research Program, Dr. Phillip Frost Department of Dermatology and Cutaneous SurgeryUniversity of Miami Miller School of MedicineMiamiFloridaUSA
- Department of Microbiology and ImmunologyUniversity of Miami Miller School of MedicineMiamiFloridaUSA
| |
Collapse
|
33
|
Qi F, Xu Y, Zheng B, Li Y, Zhang J, Liu Z, Wang X, Zhou Z, Zeng D, Lu F, Zhang C, Gan Y, Hu Z, Wang G. The Core-Shell Microneedle with Probiotic Extracellular Vesicles for Infected Wound Healing and Microbial Homeostasis Restoration. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2401551. [PMID: 39109958 DOI: 10.1002/smll.202401551] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 07/04/2024] [Indexed: 11/21/2024]
Abstract
Wound healing is a dynamic process involving the timely transition of organized phases. However, infected wounds often experience prolonged inflammation due to microbial overload. Thus, addressing the viable treatment needs across different healing stages is a critical challenge in wound management. Herein, a novel core-shell microneedle (CSMN) patch is designed for the sequential delivery of tannic acid-magnesium (TA-Mg) complexes and extracellular vesicles from Lactobacillus druckerii (LDEVs). Upon application to infected sites, CSMN@TA-Mg/LDEV releases TA-Mg first to counteract pathogenic overload and reduce reactive oxygen species (ROS), aiding the transition to proliferative phase. Subsequently, the sustained release of LDEVs enhances the activities of keratinocytes and fibroblasts, promotes vascularization, and modulates the collagen deposition. Notably, dynamic track of microbial composition demonstrates that CSMN@TA-Mg/LDEV can both inhibit the aggressive pathogen and increase the microbial diversity at wound sites. Functional analysis further highlights the potential of CSMN@TA-Mg/LDEV in facilitating wound healing and skin barrier restoration. Moreover, it is confirmed that CSMN@TA-Mg/LDEV can accelerate wound closure and improve post-recovery skin quality in the murine infected wound. Conclusively, this innovative CSMN patch offers a rapid and high-quality alternative treatment for infected wounds and emphasizes the significance of microbial homeostasis.
Collapse
Affiliation(s)
- Fangfang Qi
- Department of Plastic and Aesthetic Surgery, Nanfang Hospital of Southern Medical University, Guangzhou, 510515, China
| | - Yujie Xu
- Department of Plastic and Aesthetic Surgery, Nanfang Hospital of Southern Medical University, Guangzhou, 510515, China
| | - Bowen Zheng
- Center of Plastic and Reconstructive Surgery, Department of Plastic and Reconstructive Surgery, Zhejiang Provincial People's Hospital of Hangzhou Medical College, Hangzhou, 314408, China
| | - Yue Li
- Department of Plastic and Aesthetic Surgery, Nanfang Hospital of Southern Medical University, Guangzhou, 510515, China
| | - Jiarui Zhang
- Department of Plastic and Aesthetic Surgery, Nanfang Hospital of Southern Medical University, Guangzhou, 510515, China
| | - Zhen Liu
- Department of Plastic and Aesthetic Surgery, Nanfang Hospital of Southern Medical University, Guangzhou, 510515, China
| | - Xusheng Wang
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, 518107, China
| | - Zhiyang Zhou
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Dongqiang Zeng
- Department of Oncology, Nanfang Hospital of Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Feng Lu
- Department of Plastic and Aesthetic Surgery, Nanfang Hospital of Southern Medical University, Guangzhou, 510515, China
| | - Chunhua Zhang
- Department of Plastic and Aesthetic Surgery, Nanfang Hospital of Southern Medical University, Guangzhou, 510515, China
| | - Yuyang Gan
- Department of Plastic and Aesthetic Surgery, Nanfang Hospital of Southern Medical University, Guangzhou, 510515, China
| | - Zhiqi Hu
- Department of Plastic and Aesthetic Surgery, Nanfang Hospital of Southern Medical University, Guangzhou, 510515, China
| | - Gaofeng Wang
- Department of Plastic and Aesthetic Surgery, Nanfang Hospital of Southern Medical University, Guangzhou, 510515, China
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD, 21210, USA
| |
Collapse
|
34
|
Khadka VD, Markey L, Boucher M, Lieberman TD. Commensal Skin Bacteria Exacerbate Inflammation and Delay Skin Barrier Repair. J Invest Dermatol 2024; 144:2541-2552.e10. [PMID: 38604402 DOI: 10.1016/j.jid.2024.03.033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 03/04/2024] [Accepted: 03/16/2024] [Indexed: 04/13/2024]
Abstract
The skin microbiome can both trigger beneficial immune stimulation and pose a potential infection threat. Previous studies have shown that colonization of mouse skin with the model human skin commensal Staphylococcus epidermidis is protective against subsequent excisional wound or pathogen challenge. However, less is known about concurrent skin damage and exposure to commensal microbes, despite growing interest in interventional probiotic therapy. In this study, we address this open question by applying commensal skin bacteria at a high dose to abraded skin. Although depletion of the skin microbiome through antibiotics delayed repair from damage, probiotic-like application of commensals-including the mouse commensal Staphylococcus xylosus, 3 distinct isolates of S. epidermidis, and all other tested human skin commensals-also significantly delayed barrier repair. Increased inflammation was observed within 4 hours of S. epidermidis exposure and persisted through day 4, at which point the skin displayed a chronic wound-like inflammatory state with increased neutrophil infiltration, increased fibroblast activity, and decreased monocyte differentiation. Transcriptomic analysis suggested that the prolonged upregulation of early canonical proliferative pathways inhibited the progression of barrier repair. These results highlight the nuanced role of members of the skin microbiome in modulating barrier integrity and indicate the need for caution in their development as probiotics.
Collapse
Affiliation(s)
- Veda D Khadka
- Institute for Medical Engineering & Sciences, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA; Department of Civil and Environmental Engineering, School of Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Laura Markey
- Institute for Medical Engineering & Sciences, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA; Department of Civil and Environmental Engineering, School of Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Magalie Boucher
- The Division of Comparative Medicine, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Tami D Lieberman
- Institute for Medical Engineering & Sciences, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA; Department of Civil and Environmental Engineering, School of Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA; Ragon Institute of Mass General, Massachusetts Institute of Technology, and Harvard, Cambridge, MA, USA.
| |
Collapse
|
35
|
Zhao Z, Rong Y, Yin R, Zeng R, Xu Z, Lv D, Hu Z, Cao X, Tang B. Skin Microbiota, Immune Cell, and Skin Fibrosis: A Comprehensive Mendelian Randomization Study. Biomedicines 2024; 12:2409. [PMID: 39457721 PMCID: PMC11505207 DOI: 10.3390/biomedicines12102409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 09/25/2024] [Accepted: 10/16/2024] [Indexed: 10/28/2024] Open
Abstract
BACKGROUND Microbiota dysbiosis has been reported to lead to leaky epithelia and trigger numerous dermatological conditions. However, potential causal associations between skin microbiota and skin fibrosis and whether immune cells act as mediators remain unclear. METHODS Summary statistics of skin microbiota, immune cells, and skin fibrosis were identified from large-scale genome-wide association studies summary data. Bidirectional Mendelian randomization was performed to ascertain unidirectional causal effects between skin microbiota, immune cells, and skin fibrosis. We performed a mediation analysis to identify the role of immune cells in the pathway from skin microbiota to skin fibrosis. RESULTS Three specific skin microbiotas were positively associated with skin fibrosis, while the other three were negative. A total of 15 immune cell traits were associated with increased skin fibrosis risk, while 27 were associated with a decreased risk. Moreover, two immune cell traits were identified as mediating factors. CONCLUSIONS Causal associations were identified between skin microbiota, immune cells, and skin fibrosis. There is evidence that immune cells exert mediating effects on skin microbiota in skin fibrosis. In addition, some strains exhibit different effects on skin fibrosis in distinct environments.
Collapse
Affiliation(s)
- Zirui Zhao
- Department of Burns, Wound Repair and Reconstruction, First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China; (Z.Z.); (Y.R.); (Z.X.); (D.L.); (Z.H.)
| | - Yanchao Rong
- Department of Burns, Wound Repair and Reconstruction, First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China; (Z.Z.); (Y.R.); (Z.X.); (D.L.); (Z.H.)
| | - Rong Yin
- Department of Dermatology, First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China;
| | - Ruixi Zeng
- Department of Plastic Surgery, First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China;
| | - Zhongye Xu
- Department of Burns, Wound Repair and Reconstruction, First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China; (Z.Z.); (Y.R.); (Z.X.); (D.L.); (Z.H.)
| | - Dongming Lv
- Department of Burns, Wound Repair and Reconstruction, First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China; (Z.Z.); (Y.R.); (Z.X.); (D.L.); (Z.H.)
| | - Zhicheng Hu
- Department of Burns, Wound Repair and Reconstruction, First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China; (Z.Z.); (Y.R.); (Z.X.); (D.L.); (Z.H.)
| | - Xiaoling Cao
- Department of Burns, Wound Repair and Reconstruction, First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China; (Z.Z.); (Y.R.); (Z.X.); (D.L.); (Z.H.)
| | - Bing Tang
- Department of Burns, Wound Repair and Reconstruction, First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China; (Z.Z.); (Y.R.); (Z.X.); (D.L.); (Z.H.)
| |
Collapse
|
36
|
Radaschin DS, Tatu A, Iancu AV, Beiu C, Popa LG. The Contribution of the Skin Microbiome to Psoriasis Pathogenesis and Its Implications for Therapeutic Strategies. MEDICINA (KAUNAS, LITHUANIA) 2024; 60:1619. [PMID: 39459406 PMCID: PMC11509136 DOI: 10.3390/medicina60101619] [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: 09/17/2024] [Revised: 09/26/2024] [Accepted: 09/30/2024] [Indexed: 10/28/2024]
Abstract
Psoriasis is a common chronic inflammatory skin disease, associated with significant morbidity and a considerable negative impact on the patients' quality of life. The complex pathogenesis of psoriasis is still incompletely understood. Genetic predisposition, environmental factors like smoking, alcohol consumption, psychological stress, consumption of certain drugs, and mechanical trauma, as well as specific immune dysfunctions, contribute to the onset of the disease. Mounting evidence indicate that skin dysbiosis plays a significant role in the development and exacerbation of psoriasis through loss of immune tolerance to commensal skin flora, an altered balance between Tregs and effector cells, and an excessive Th1 and Th17 polarization. While the implications of skin dysbiosis in psoriasis pathogenesis are only starting to be revealed, the progress in the characterization of the skin microbiome changes in psoriasis patients has opened a whole new avenue of research focusing on the modulation of the skin microbiome as an adjuvant treatment for psoriasis and as part of a long-term plan to prevent disease flares. The skin microbiome may also represent a valuable predictive marker of treatment response and may aid in the selection of the optimal personalized treatment. We present the current knowledge on the skin microbiome changes in psoriasis and the results of the studies that investigated the efficacy of the different skin microbiome modulation strategies in the management of psoriasis, and discuss the complex interaction between the host and skin commensal flora.
Collapse
Affiliation(s)
- Diana Sabina Radaschin
- Department of Clinical Medical, Faculty of Medicine and Pharmacy, “Saint Parascheva” Infectious Disease Clinical Hospital, Multidisciplinary Integrated Centre of Dermatological Interface Research Centre (MICDIR), “Dunarea de Jos” University of Galati, 800008 Galati, Romania
| | - Alin Tatu
- Department of Clinical Medical, Faculty of Medicine and Pharmacy, “Saint Parascheva” Infectious Disease Clinical Hospital, Multidisciplinary Integrated Centre of Dermatological Interface Research Centre (MICDIR), “Dunarea de Jos” University of Galati, 800008 Galati, Romania
| | - Alina Viorica Iancu
- Department of Morphological and Functional Sciences, “Dunarea de Jos” University of Galati, 800008 Galati, Romania
| | - Cristina Beiu
- Department of Oncologic Dermatology, Elias Emergency University Hospital, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania
| | - Liliana Gabriela Popa
- Department of Oncologic Dermatology, Elias Emergency University Hospital, Carol Davila University of Medicine and Pharmacy, 020021 Bucharest, Romania
| |
Collapse
|
37
|
Piazzesi A, Scanu M, Ciprandi G, Putignani L. Modulations of the skin microbiome in skin disorders: A narrative review from a wound care perspective. Int Wound J 2024; 21:e70087. [PMID: 39379177 PMCID: PMC11461044 DOI: 10.1111/iwj.70087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 09/19/2024] [Accepted: 09/19/2024] [Indexed: 10/10/2024] Open
Abstract
The cutaneous microbiome represents a highly dynamic community of bacteria, fungi and viruses. Scientific evidence, particularly from the last two decades, has revealed that these organisms are far from being inconsequential microscopic hitchhikers on the human body, nor are they all opportunistic pathogens waiting for the chance to penetrate the skin barrier and cause infection. In this review, we will describe how dermatological diseases have been found to be associated with disruptions and imbalances in the skin microbiome and how this new evidence had shaped the diagnosis and clinical practice relating to these disorders. We will identify the microbial agents which have been found to directly exacerbate skin diseases, as well as those which can ameliorate many of the symptoms associated with dermatological disorders. Furthermore, we will discuss the studies which suggest that bacteriotherapy, either by topical use of probiotics or by bacteria-derived compounds, can rectify skin microbial imbalances, thereby offering a promising alternative to antibiotic treatment and reducing the risks of antibiotic resistance.
Collapse
Affiliation(s)
- Antonia Piazzesi
- Immunology, Rheumatology and Infectious Diseases Research Unit, Unit of the MicrobiomeBambino Gesù Children's Hospital, IRCCSRomeItaly
| | - Matteo Scanu
- Immunology, Rheumatology and Infectious Diseases Research Unit, Unit of the MicrobiomeBambino Gesù Children's Hospital, IRCCSRomeItaly
| | - Guido Ciprandi
- Research Institute Division of Plastic and Maxillofacial Surgery, Department of SurgeryBambino Gesu' Children's Hospital, IRCCSRomeItaly
| | - Lorenza Putignani
- Unit of Microbiology and Diagnostic Immunology, Unit of Microbiomics; and Immunology, Rheumatology and Infectious Diseases Research Unit, Unit of the MicrobiomeBambino Gesù Children's Hospital, IRCCSRomeItaly
| |
Collapse
|
38
|
Radaschin DS, Iancu AV, Ionescu AM, Gurau G, Niculet E, Bujoreanu FC, Beiu C, Tatu AL, Popa LG. Comparative Analysis of the Cutaneous Microbiome in Psoriasis Patients and Healthy Individuals-Insights into Microbial Dysbiosis: Final Results. Int J Mol Sci 2024; 25:10583. [PMID: 39408916 PMCID: PMC11477231 DOI: 10.3390/ijms251910583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 09/25/2024] [Accepted: 09/29/2024] [Indexed: 10/20/2024] Open
Abstract
Psoriasis is one of the most frequent chronic inflammatory skin diseases and exerts a significant psychological impact, causing stigmatization, low self-esteem and depression. The pathogenesis of psoriasis is remarkably complex, involving genetic, immune and environmental factors, some of which are still incompletely explored. The cutaneous microbiome has become more and more important in the pathogenesis of inflammatory skin diseases such as acne, rosacea, atopic dermatitis and psoriasis. Dysbiosis of the skin microbiome could be linked to acute flare ups in psoriatic disease, as recent studies suggest. Given this hypothesis, we conducted a study in which we evaluated the cutaneous microbiome of psoriasis patients and healthy individuals. In our study, we collected multiple samples using swab sampling, adhesive tape and punch biopsies. Our results are similar to other studies in which the qualitative and quantitative changes found in the cutaneous microbiome of psoriasis patients are different than healthy individuals. Larger, standardized studies are needed in order to elucidate the microbiome changes in psoriasis patients, clarify their role in the pathogenesis of psoriasis, decipher the interactions between the commensal microorganisms of the same and different niches and between microbiomes and the host and identify new therapeutic strategies.
Collapse
Affiliation(s)
- Diana Sabina Radaschin
- Department of Dermatology, “Saint Parascheva” Infectious Disease Clinical Hospital, 800179 Galati, Romania; (D.S.R.); (F.C.B.)
- Department of Clinical Medical, Faculty of Medicine and Pharmacy, “Dunarea de Jos” University, 800008 Galati, Romania
- Multidisciplinary Integrated Centre of Dermatological Interface Research Centre (MICDIR), “Dunarea de Jos” University, 800008 Galati, Romania
| | - Alina Viorica Iancu
- Department of Morphological and Functional Sciences, Faculty of Medicine and Pharmacy, “Dunarea de Jos” University, 800008 Galati, Romania; (A.V.I.); (G.G.); (E.N.)
| | | | - Gabriela Gurau
- Department of Morphological and Functional Sciences, Faculty of Medicine and Pharmacy, “Dunarea de Jos” University, 800008 Galati, Romania; (A.V.I.); (G.G.); (E.N.)
| | - Elena Niculet
- Department of Morphological and Functional Sciences, Faculty of Medicine and Pharmacy, “Dunarea de Jos” University, 800008 Galati, Romania; (A.V.I.); (G.G.); (E.N.)
| | - Florin Ciprian Bujoreanu
- Department of Dermatology, “Saint Parascheva” Infectious Disease Clinical Hospital, 800179 Galati, Romania; (D.S.R.); (F.C.B.)
- Department of Clinical Medical, Faculty of Medicine and Pharmacy, “Dunarea de Jos” University, 800008 Galati, Romania
- Multidisciplinary Integrated Centre of Dermatological Interface Research Centre (MICDIR), “Dunarea de Jos” University, 800008 Galati, Romania
| | - Cristina Beiu
- Dermatology Department, Carol Davila University of Medicine and Pharmacy, 030167 Bucharest, Romania;
| | - Alin Laurentiu Tatu
- Department of Dermatology, “Saint Parascheva” Infectious Disease Clinical Hospital, 800179 Galati, Romania; (D.S.R.); (F.C.B.)
- Department of Clinical Medical, Faculty of Medicine and Pharmacy, “Dunarea de Jos” University, 800008 Galati, Romania
- Multidisciplinary Integrated Centre of Dermatological Interface Research Centre (MICDIR), “Dunarea de Jos” University, 800008 Galati, Romania
| | - Liliana Gabriela Popa
- Dermatology Department, Carol Davila University of Medicine and Pharmacy, 030167 Bucharest, Romania;
| |
Collapse
|
39
|
Manus MB, Savo Sardaro ML, Dada O, Davis M, Romoff MR, Torello SG, Ubadigbo E, Wu RC, Dominguez-Bello MG, Melby MK, Miller ES, Amato KR. Birth and household exposures are associated with changes to skin bacterial communities during infancy. Evol Med Public Health 2024; 13:49-76. [PMID: 40182701 PMCID: PMC11966193 DOI: 10.1093/emph/eoae023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Revised: 08/01/2024] [Indexed: 04/05/2025] Open
Abstract
Background and objectives Microbial exposures during infancy shape the development of the microbiome, the collection of microbes living in and on the body, which in turn directs immune system training. Newborns acquire a substantial quantity of microbes during birth and throughout infancy via exposure to microbes in the physical and social environment. Alterations to early life microbial environments may give rise to mismatches, where environmental, cultural and behavioral changes that outpace the body's adaptive responses can lead to adverse health outcomes, particularly those related to microbiome development and immune system regulation. Methods This study explored the development of the skin microbiome among infants born in Chicago, USA. We collected skin swab microbiome samples from 22 mother-infant dyads during the first 48 h of life and again at 6 weeks postpartum. Mothers provided information about social environments and hygiene behaviors that may impact infants' microbial exposures. Results Analysis of amplicon bacterial gene sequencing data revealed correlations between infant skin bacterial abundances shortly after birth and factors such as antibiotic exposure and receiving a bath in the hospital. The composition of the infant microbiome at 6 weeks of age was associated with interactions with caregivers and infant feeding practices. We also found shifts in maternal skin microbiomes that may reflect increased hygiene practices in the hospital. Conclusions and implications Our data suggest that factors related to the birth and household environment can impact the development of infant skin microbiomes and point to practices that may produce mismatches for the infant microbiome and immune system.
Collapse
Affiliation(s)
- Melissa B Manus
- Department of Anthropology, University of Texas at San Antonio, San Antonio, TX, USA
- Department of Anthropology, Northwestern University, Evanston, IL, USA
| | - Maria Luisa Savo Sardaro
- Department of Anthropology, Northwestern University, Evanston, IL, USA
- Department of Human Science and Promotion of the Quality of Life, University of San Raffaele, Rome, Italy
| | - Omolola Dada
- Department of Anthropology, Northwestern University, Evanston, IL, USA
| | - Maya Davis
- Department of Anthropology, Northwestern University, Evanston, IL, USA
| | - Melissa R Romoff
- Department of Anthropology, Northwestern University, Evanston, IL, USA
| | | | - Esther Ubadigbo
- Department of Anthropology, Northwestern University, Evanston, IL, USA
| | - Rebecca C Wu
- Department of Anthropology, Northwestern University, Evanston, IL, USA
| | - Maria Gloria Dominguez-Bello
- Department of Biochemistry and Microbiology, Rutgers University, New Brunswick, NJ, USA
- Department of Anthropology, Rutgers University, New Brunswick, NJ, USA
- Humans and the Microbiome Program, Canadian Institute for Advanced Research, Toronto, ON, Canada
| | - Melissa K Melby
- Department of Anthropology, University of Delaware, Newark, DE, USA
- Humans and the Microbiome Program, Canadian Institute for Advanced Research, Toronto, ON, Canada
| | - Emily S Miller
- Department of Obstetrics and Gynecology, Division of Maternal Fetal Medicine, Warren Alpert Medical School of Brown University, Providence, RI;USA
| | - Katherine R Amato
- Department of Anthropology, Northwestern University, Evanston, IL, USA
- Humans and the Microbiome Program, Canadian Institute for Advanced Research, Toronto, ON, Canada
| |
Collapse
|
40
|
Christman L, De Benedetto A, Johnson E, Khoo C, Gu L. Polyphenol-Rich Cranberry Beverage Positively Affected Skin Health, Skin Lipids, Skin Microbiome, Inflammation, and Oxidative Stress in Women in a Randomized Controlled Trial. Nutrients 2024; 16:3126. [PMID: 39339726 PMCID: PMC11434900 DOI: 10.3390/nu16183126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 08/27/2024] [Accepted: 08/29/2024] [Indexed: 09/30/2024] Open
Abstract
This study aimed to determine whether a polyphenol-rich cranberry beverage affects skin properties, lipids, and the microbiome in women using a randomized, double-blinded, placebo-controlled, cross-over design. Twenty-two women with Fitzpatrick skin types 2-3 were randomized to drink a cranberry beverage or placebo for six weeks. After a 21-day washout, they consumed the opposite beverage for six weeks. Six weeks of cranberry beverage significantly reduced UVB-induced erythema, improved net elasticity on the face and forearm, smoothness on the face, and gross elasticity on the forearm compared to the placebo. When stratified by age, these effects of the cranberry beverage were primarily observed in women >40 years old. SOD activities were improved after six weeks of cranberry beverage consumption compared to the placebo, while glutathione peroxide and TNF-α were improved compared to baseline. These effects were found to differ by age group. Skin lipid composition was modulated by both the cranberry beverage and the placebo. Cranberry beverages did not change α- or β-diversity but altered the abundance of several skin microbes at the species and strain level. Consumption of a cranberry beverage for six weeks improved specific skin properties and oxidative stress and modulated skin lipids and microbiome compared to placebo.
Collapse
Affiliation(s)
- Lindsey Christman
- Food Science and Human Nutrition Department, Institute of Food and Agricultural Sciences, University of Florida, Gainesville, FL 32611, USA
| | - Anna De Benedetto
- Department of Dermatology, College of Medicine, University of Florida, Gainesville, FL 32611, USA
- Department of Dermatology, University of Rochester Medical Center, Rochester, NY 14620, USA
| | | | | | - Liwei Gu
- Food Science and Human Nutrition Department, Institute of Food and Agricultural Sciences, University of Florida, Gainesville, FL 32611, USA
| |
Collapse
|
41
|
Sun C, Hu G, Yi L, Ge W, Yang Q, Yang X, He Y, Liu Z, Chen WH. Integrated analysis of facial microbiome and skin physio-optical properties unveils cutotype-dependent aging effects. MICROBIOME 2024; 12:163. [PMID: 39232827 PMCID: PMC11376020 DOI: 10.1186/s40168-024-01891-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Accepted: 07/29/2024] [Indexed: 09/06/2024]
Abstract
BACKGROUND Our facial skin hosts millions of microorganisms, primarily bacteria, crucial for skin health by maintaining the physical barrier, modulating immune response, and metabolizing bioactive materials. Aging significantly influences the composition and function of the facial microbiome, impacting skin immunity, hydration, and inflammation, highlighting potential avenues for interventions targeting aging-related facial microbes amidst changes in skin physiological properties. RESULTS We conducted a multi-center and deep sequencing survey to investigate the intricate interplay of aging, skin physio-optical conditions, and facial microbiome. Leveraging a newly-generated dataset of 2737 species-level metagenome-assembled genomes (MAGs), our integrative analysis highlighted aging as the primary driver, influencing both facial microbiome composition and key skin characteristics, including moisture, sebum production, gloss, pH, elasticity, and sensitivity. Further mediation analysis revealed that skin characteristics significantly impacted the microbiome, mostly as a mediator of aging. Utilizing this dataset, we uncovered two consistent cutotypes across sampling cities and identified aging-related microbial MAGs. Additionally, a Facial Aging Index (FAI) was formulated based on the microbiome, uncovering the cutotype-dependent effects of unhealthy lifestyles on skin aging. Finally, we distinguished aging related microbial pathways influenced by lifestyles with cutotype-dependent effect. CONCLUSIONS Together, our findings emphasize aging's central role in facial microbiome dynamics, and support personalized skin microbiome interventions by targeting lifestyle, skin properties, and aging-related microbial factors. Video Abstract.
Collapse
Affiliation(s)
- Chuqing Sun
- Department of Bioinformatics and Systems Biology, College of Life Science and Technology, Key Laboratory of Molecular Biophysics of the Ministry of Education, Hubei Key Laboratory of Bioinformatics and Molecular Imaging, Center for Artificial Intelligence Biology, Huazhong University of Science and Technology, Wuhan, 430074, China
- Center for Research and Development, Xiamen Treatgut Biotechnology Co., Ltd., Xiamen, China
- School of Life Sciences, Xiamen University, Xiamen, China
| | - Guoru Hu
- Department of Bioinformatics and Systems Biology, College of Life Science and Technology, Key Laboratory of Molecular Biophysics of the Ministry of Education, Hubei Key Laboratory of Bioinformatics and Molecular Imaging, Center for Artificial Intelligence Biology, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Liwen Yi
- Department of Biotechnology, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Wei Ge
- Department of Dermatology, Huazhong University of Science and Technology Hospital, Wuhan, 430074, China
| | - Qingyu Yang
- Department of Dermatology, Huazhong University of Science and Technology Hospital, Wuhan, 430074, China
| | - Xiangliang Yang
- Department of Biotechnology, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
- College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
- National Engineering Research Center for Nanomedicine, Wuhan, 430074, China
| | - Yifan He
- The GBA National Institute for Nanotechnology Innovation, Guangzhou, 510799, China.
- School of Biomedical Science and Engineering, South China University of Technology, Guangzhou, 510641, China.
- College of Chemistry and Materials Engineering and Institute of Cosmetic Regulatory Science, Beijing Technology and Business University, Beijing, 100048, People's Republic of China.
| | - Zhi Liu
- Department of Biotechnology, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China.
| | - Wei-Hua Chen
- Department of Bioinformatics and Systems Biology, College of Life Science and Technology, Key Laboratory of Molecular Biophysics of the Ministry of Education, Hubei Key Laboratory of Bioinformatics and Molecular Imaging, Center for Artificial Intelligence Biology, Huazhong University of Science and Technology, Wuhan, 430074, China.
- Institution of Medical Artificial Intelligence, Binzhou Medical University, Yantai, 264003, China.
| |
Collapse
|
42
|
Berdyshev E. Skin Lipid Barrier: Structure, Function and Metabolism. ALLERGY, ASTHMA & IMMUNOLOGY RESEARCH 2024; 16:445-461. [PMID: 39363765 PMCID: PMC11450438 DOI: 10.4168/aair.2024.16.5.445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Revised: 08/12/2024] [Accepted: 08/13/2024] [Indexed: 10/05/2024]
Abstract
Lipids are important skin components that provide, together with proteins, barrier function of the skin. Keratinocyte terminal differentiation launches unique metabolic changes to lipid metabolism that result in the predominance of ceramides within lipids of the stratum corneum (SC)-the very top portion of the skin. Differentiating keratinocytes form unique ceramides that can be found only in the skin, and generate specialized extracellular structures known as lamellae. Lamellae establish tight hydrophobic layers between dying keratinocytes to protect the body from water loss and also from penetration of allergens and bacteria. Genetic and immunological factors may lead to the failure of keratinocyte terminal differentiation and significantly alter the proportion between SC components. The consequence of such changes is loss or deterioration of skin barrier function that can lead to pathological changes in the skin. This review summarizes our current understanding of the role of lipids in skin barrier function. It also draws attention to the utility of testing SC for lipid and protein biomarkers to predict future onset of allergic skin diseases.
Collapse
Affiliation(s)
- Evgeny Berdyshev
- Department of Medicine, National Jewish Health, Denver, CO, USA.
| |
Collapse
|
43
|
Rühling M, Kersting L, Wagner F, Schumacher F, Wigger D, Helmerich DA, Pfeuffer T, Elflein R, Kappe C, Sauer M, Arenz C, Kleuser B, Rudel T, Fraunholz M, Seibel J. Trifunctional sphingomyelin derivatives enable nanoscale resolution of sphingomyelin turnover in physiological and infection processes via expansion microscopy. Nat Commun 2024; 15:7456. [PMID: 39198435 PMCID: PMC11358447 DOI: 10.1038/s41467-024-51874-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Accepted: 08/20/2024] [Indexed: 09/01/2024] Open
Abstract
Sphingomyelin is a key molecule of sphingolipid metabolism, and its enzymatic breakdown is associated with various infectious diseases. Here, we introduce trifunctional sphingomyelin derivatives that enable the visualization of sphingomyelin distribution and sphingomyelinase activity in infection processes. We demonstrate this by determining the activity of a bacterial sphingomyelinase on the plasma membrane of host cells using a combination of Förster resonance energy transfer and expansion microscopy. We further use our trifunctional sphingomyelin probes to visualize their metabolic state during infections with Chlamydia trachomatis and thereby show that chlamydial inclusions primarily contain the cleaved forms of the molecules. Using expansion microscopy, we observe that the proportion of metabolized molecules increases during maturation from reticulate to elementary bodies, indicating different membrane compositions between the two chlamydial developmental forms. Expansion microscopy of trifunctional sphingomyelins thus provides a powerful microscopy tool to analyze sphingomyelin metabolism in cells at nanoscale resolution.
Collapse
Affiliation(s)
- Marcel Rühling
- Chair of Microbiology, Julius-Maximilians-University Würzburg, Würzburg, Germany
| | - Louise Kersting
- Institute of Organic Chemistry, Julius-Maximilians-University Würzburg, Würzburg, Germany
| | - Fabienne Wagner
- Chair of Microbiology, Julius-Maximilians-University Würzburg, Würzburg, Germany
| | | | - Dominik Wigger
- Institute of Pharmacy, Freie Universität Berlin, Berlin, Germany
| | - Dominic A Helmerich
- Chair of Biotechnology & Biophysics, Biocenter, Julius-Maximilians-University Würzburg, Würzburg, Germany
| | - Tom Pfeuffer
- Institute of Organic Chemistry, Julius-Maximilians-University Würzburg, Würzburg, Germany
| | - Robin Elflein
- Institute of Organic Chemistry, Julius-Maximilians-University Würzburg, Würzburg, Germany
| | - Christian Kappe
- Institute of Chemistry, Humboldt-Universität zu Berlin, Brook-Taylor-Str 2, Berlin, Germany
| | - Markus Sauer
- Chair of Biotechnology & Biophysics, Biocenter, Julius-Maximilians-University Würzburg, Würzburg, Germany
| | - Christoph Arenz
- Institute of Chemistry, Humboldt-Universität zu Berlin, Brook-Taylor-Str 2, Berlin, Germany
| | - Burkhard Kleuser
- Institute of Pharmacy, Freie Universität Berlin, Berlin, Germany
| | - Thomas Rudel
- Chair of Microbiology, Julius-Maximilians-University Würzburg, Würzburg, Germany
| | - Martin Fraunholz
- Chair of Microbiology, Julius-Maximilians-University Würzburg, Würzburg, Germany
| | - Jürgen Seibel
- Institute of Organic Chemistry, Julius-Maximilians-University Würzburg, Würzburg, Germany.
| |
Collapse
|
44
|
Jo A, Kim KS, Won J, Shin H, Kim S, Kim B, Kim DJ, Cho JY, Kim HJ. Nasal symbiont Staphylococcus epidermidis restricts influenza A virus replication via the creation of a polyamine-deficient cellular environment. Commun Biol 2024; 7:1031. [PMID: 39174732 PMCID: PMC11341892 DOI: 10.1038/s42003-024-06706-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Accepted: 08/08/2024] [Indexed: 08/24/2024] Open
Abstract
Studies on the immune-regulatory roles played by the commensal microbes residing in the nasal mucosa consider the contribution of antiviral immune responses. Here, we sought to identify the nasal microbiome, Staphylococcus epidermidis-regulated antiviral immune responses and the alteration of polyamine metabolites in nasal epithelium. We found that polyamines were required for the life cycle of influenza A virus (IAV) and depletion of polyamines disturbed IAV replication in normal human nasal epithelial (NHNE) cells. Inoculation of S. epidermidis also suppressed IAV infection and the concentration of polyamines including putrescine, spermidine, and spermine was completely attenuated in S. epidermidis-inoculated NHNE cells. S. epidermidis activated the enzyme involved in the production of ornithine from arginine and downregulated the activity of the enzyme involved in the production of putrescine from ornithine in nasal epithelium. S. epidermidis also induced the activation of enzymes that promote the extracellular export of spermine and spermidine in NHNE cells. Our findings demonstrate that S. epidermidis is shown to be able of creating an intracellular environment lacking polyamines in the nasal epithelium and promote the balance of cellular polyamines in favor of the host to restrict influenza virus replication.
Collapse
Affiliation(s)
- Ara Jo
- Department of Otorhinolaryngology, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Kyeong-Seog Kim
- Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine and Hospital, Seoul, Republic of Korea
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Jina Won
- Department of Otorhinolaryngology, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Haeun Shin
- Department of Otorhinolaryngology, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Sujin Kim
- Department of Otorhinolaryngology, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Bora Kim
- Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine and Hospital, Seoul, Republic of Korea
- Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Da Jung Kim
- Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine and Hospital, Seoul, Republic of Korea
- Liver Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Joo-Youn Cho
- Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine and Hospital, Seoul, Republic of Korea.
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Republic of Korea.
| | - Hyun Jik Kim
- Department of Otorhinolaryngology, Seoul National University College of Medicine, Seoul, Republic of Korea.
- Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine and Hospital, Seoul, Republic of Korea.
- Seoul National University Hospital, Seoul, Republic of Korea.
- Sensory Organ Research Institute, Seoul National University Medical Research Center, Seoul, Republic of Korea.
| |
Collapse
|
45
|
Pagac MP, Gempeler M, Campiche R. A New Generation of Postbiotics for Skin and Scalp: In Situ Production of Lipid Metabolites by Malassezia. Microorganisms 2024; 12:1711. [PMID: 39203553 PMCID: PMC11357556 DOI: 10.3390/microorganisms12081711] [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: 07/10/2024] [Revised: 07/24/2024] [Accepted: 07/31/2024] [Indexed: 09/03/2024] Open
Abstract
Effects of pre- and probiotics on intestinal health are well researched and microbiome-targeting solutions are commercially available. Even though a trend to appreciate the presence of certain microbes on the skin is seeing an increase in momentum, our understanding is limited as to whether the utilization of skin-resident microbes for beneficial effects holds the same potential as the targeted manipulation of the gut microflora. Here, we present a selection of molecular mechanisms of cross-communication between human skin and the skin microbial community and the impact of these interactions on the host's cutaneous health with implications for the development of skin cosmetic and therapeutic solutions. Malassezia yeasts, as the main fungal representatives of the skin microfloral community, interact with the human host skin via lipid mediators, of which several are characterized by exhibiting potent anti-inflammatory activities. This review therefore puts a spotlight on Malassezia and provides a comprehensive overview of the current state of knowledge about these fungal-derived lipid mediators and their capability to reduce aesthetical and sensory burdens, such as redness and itching, commonly associated with inflammatory skin conditions. Finally, several examples of current skin microbiome-based interventions for cosmetic solutions are discussed, and models are presented for the use of skin-resident microbes as endogenous bio-manufacturing platforms for the in situ supplementation of the skin with beneficial metabolites.
Collapse
Affiliation(s)
- Martin Patrick Pagac
- DSM-Firmenich, Perfumery & Beauty, Wurmisweg 576, CH-4303 Kaiseraugst, Switzerland; (M.G.); (R.C.)
| | | | | |
Collapse
|
46
|
MacGibeny MA, Adjei S, Pyle H, Bunick CG, Ghannoum M, Grada A, Harris-Tryon T, Tyring SK, Kong HH. The Human Skin Microbiome in Health: CME Part 1. J Am Acad Dermatol 2024:S0190-9622(24)02671-9. [PMID: 39168311 PMCID: PMC11912297 DOI: 10.1016/j.jaad.2024.07.1498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Revised: 06/15/2024] [Accepted: 07/03/2024] [Indexed: 08/23/2024]
Abstract
Human skin is home to a myriad of microorganisms, including bacteria, viruses, fungi, and mites, many of which are considered commensal microbes that aid in maintaining the overall homeostasis or steady-state condition of the skin and contribute to skin health. Our understanding of the complexities of the skin's interaction with its microorganisms is evolving. This knowledge is based primarily on in vitro and animal studies, and more work is needed to understand how this knowledge relates to humans. Here, we introduce the concept of the skin microbiome and discuss skin microbial ecology, some intrinsic factors with potential influence on the human skin microbiome, and possible microbiome-host interactions. The second article of this two-part CME series describes how microbiome alterations may be associated with skin disease, how medications can affect the microbiome, and what microbiome-based therapies are under investigation.
Collapse
Affiliation(s)
| | - Susuana Adjei
- Department of Dermatology, Lake Granbury Medical Center, Dallas, TX, USA
| | - Hunter Pyle
- Department of Dermatology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Christopher G Bunick
- Department of Dermatology, Yale School of Medicine, New Haven, CT, USA; Program in Translational Biomedicine, Yale School of Medicine, New Haven, CT, USA
| | - Mahmoud Ghannoum
- Integrated Microbiome Core and Center for Medical Mycology, Case Western Reserve University and University Hospitals Cleveland Medical Center, Cleveland, OH, USA; Department of Dermatology, Case Western Reserve University and University Hospitals Cleveland Medical Center, Cleveland, OH, USA
| | - Ayman Grada
- Integrated Microbiome Core and Center for Medical Mycology, Case Western Reserve University and University Hospitals Cleveland Medical Center, Cleveland, OH, USA
| | - Tamia Harris-Tryon
- Department of Dermatology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Stephen K Tyring
- Department of Dermatology, Lake Granbury Medical Center, Dallas, TX, USA.
| | - Heidi H Kong
- Dermatology Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
47
|
Zhang X, Huang X, Zheng P, Liu E, Bai S, Chen S, Pang Y, Xiao X, Yang H, Guo J. Changes in oral, skin, and gut microbiota in children with atopic dermatitis: a case-control study. Front Microbiol 2024; 15:1442126. [PMID: 39211320 PMCID: PMC11358084 DOI: 10.3389/fmicb.2024.1442126] [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: 06/01/2024] [Accepted: 07/31/2024] [Indexed: 09/04/2024] Open
Abstract
Introduction Atopic dermatitis (AD) is a common clinical recurrent atopic disease in dermatology, most seen in children and adolescents. In recent years, AD has been found to be closely associated with microbial communities. Methods To explore the synergistic effects between colonizing bacteria from different sites and AD, we comparatively analyzed the skin, oral, and gut microbiota of children with AD (50 individuals) and healthy children (50 individuals) by 16S rRNA gene sequencing. Twenty samples were also randomly selected from both groups for metabolic and macrogenomic sequencing. Results The results of our sequencing study showed reduced microbiota diversity in the oral, skin, and gut of children with AD (P < 0.05). Metabolomics analysis showed that serotonergic synapse, arachidonic acid metabolism, and steroid biosynthesis were downregulated at all three loci in the oral, skin, and gut of children with AD (P < 0.05). Macrogenomic sequencing analysis showed that KEGG functional pathways of the three site flora were involved in oxidative phosphorylation, ubiquitin-mediated proteolysis, mRNA surveillance pathway, ribosome biogenesis in eukaryotes, proteasome, basal transcription factors, peroxisome, MAPK signaling pathway, mitophagy, fatty acid elongation, and so on (P < 0.05). Discussion The combined microbial, metabolic, and macrogenetic analyses identified key bacteria, metabolites, and pathogenic pathways that may be associated with AD development. We provides a more comprehensive and in-depth understanding of the role of the microbiota at different sites in AD patients, pointing to new directions for future diagnosis, treatment and prognosis.
Collapse
Affiliation(s)
- Xueer Zhang
- Department of Dermatology, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xiaomin Huang
- Department of Dermatology, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Pai Zheng
- Department of Dermatology, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - E. Liu
- Department of Dermatology, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Sixian Bai
- Department of Dermatology, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Shuoyu Chen
- Department of Dermatology, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yaobin Pang
- Department of Dermatology, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xinyu Xiao
- Department of Dermatology, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Huifang Yang
- Department of Dermatology, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jing Guo
- Department of Dermatology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
48
|
Gan Y, Zhang J, Qi F, Hu Z, Sweren E, Reddy SK, Chen L, Feng X, Grice EA, Garza LA, Wang G. Commensal microbe regulation of skin cells in disease. Cell Host Microbe 2024; 32:1264-1279. [PMID: 39146798 PMCID: PMC11457753 DOI: 10.1016/j.chom.2024.07.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 07/18/2024] [Accepted: 07/19/2024] [Indexed: 08/17/2024]
Abstract
Human skin is the host to various commensal microbes that constitute a substantial microbial community. The reciprocal communication between these microbial inhabitants and host cells upholds both the morphological and functional attributes of the skin layers, contributing indispensably to microenvironmental and tissue homeostasis. Thus, disruption of the skin barrier or imbalances in the microbial communities can exert profound effects on the behavior of host cells. This influence, mediated by the microbes themselves or their metabolites, manifests in diverse outcomes. In this review, we examine existing knowledge to provide insight into the nuanced behavior exhibited by the microbiota on skin cells in health and disease states. These interactions provide insight into potential cellular targets for future microbiota-based therapies to prevent and treat skin disease.
Collapse
Affiliation(s)
- Yuyang Gan
- Department of Plastic and Aesthetic Surgery, Nanfang Hospital of Southern Medical University, Guangzhou, Guangdong Province 510515, China
| | - Jiarui Zhang
- Department of Plastic and Aesthetic Surgery, Nanfang Hospital of Southern Medical University, Guangzhou, Guangdong Province 510515, China
| | - Fangfang Qi
- Department of Plastic and Aesthetic Surgery, Nanfang Hospital of Southern Medical University, Guangzhou, Guangdong Province 510515, China
| | - Zhiqi Hu
- Department of Plastic and Aesthetic Surgery, Nanfang Hospital of Southern Medical University, Guangzhou, Guangdong Province 510515, China
| | - Evan Sweren
- University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Sashank K Reddy
- Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD 21210, USA; Department of Plastic and Reconstructive Surgery, Johns Hopkins School of Medicine, Baltimore, MD 21287, USA
| | - Lu Chen
- Department of Plastic and Aesthetic Surgery, Nanfang Hospital of Southern Medical University, Guangzhou, Guangdong Province 510515, China
| | - Xinyi Feng
- Department of Plastic and Aesthetic Surgery, Nanfang Hospital of Southern Medical University, Guangzhou, Guangdong Province 510515, China
| | - Elizabeth A Grice
- Department of Dermatology and Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| | - Luis A Garza
- Department of Plastic and Aesthetic Surgery, Nanfang Hospital of Southern Medical University, Guangzhou, Guangdong Province 510515, China; Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD 21210, USA; Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21210, USA.
| | - Gaofeng Wang
- Department of Plastic and Aesthetic Surgery, Nanfang Hospital of Southern Medical University, Guangzhou, Guangdong Province 510515, China; Department of Dermatology, Johns Hopkins University School of Medicine, Baltimore, MD 21210, USA.
| |
Collapse
|
49
|
Locker J, Serrage HJ, Ledder RG, Deshmukh S, O'Neill CA, McBain AJ. Microbiological insights and dermatological applications of live biotherapeutic products. J Appl Microbiol 2024; 135:lxae181. [PMID: 39090975 DOI: 10.1093/jambio/lxae181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 05/26/2024] [Accepted: 07/31/2024] [Indexed: 08/04/2024]
Abstract
As our understanding of dermatological conditions advances, it becomes increasingly evident that traditional pharmaceutical interventions are not universally effective. The intricate balance of the skin microbiota plays a pivotal role in the development of various skin conditions, prompting a growing interest in probiotics, or live biotherapeutic products (LBPs), as potential remedies. Specifically, the topical application of LBPs to modulate bacterial populations on the skin has emerged as a promising approach to alleviate symptoms associated with common skin conditions. This review considers LBPs and their application in addressing a wide spectrum of dermatological conditions with particular emphasis on three key areas: acne, atopic dermatitis, and wound healing. Within this context, the critical role of strain selection is presented as a pivotal factor in effectively managing these dermatological concerns. Additionally, the review considers formulation challenges associated with probiotic viability and proposes a personalised approach to facilitate compatibility with the skin's unique microenvironment. This analysis offers valuable insights into the potential of LBPs in dermatological applications, underlining their promise in reshaping the landscape of dermatological treatments while acknowledging the hurdles that must be overcome to unlock their full potential.
Collapse
Affiliation(s)
- Jessica Locker
- Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, M13 9PT, UK
| | - Hannah J Serrage
- Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, M13 9PT, UK
- Division of Musculoskeletal and Dermatological Science, Faculty of Biology, Medicine and Health, School of Biological Science, The University of Manchester, Manchester, M13 9PT, UK
| | - Ruth G Ledder
- Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, M13 9PT, UK
| | | | - Catherine A O'Neill
- Division of Musculoskeletal and Dermatological Science, Faculty of Biology, Medicine and Health, School of Biological Science, The University of Manchester, Manchester, M13 9PT, UK
| | - Andrew J McBain
- Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, M13 9PT, UK
| |
Collapse
|
50
|
D’Arcangelo S, Di Fermo P, Diban F, Ferrone V, D’Ercole S, Di Giulio M, Di Lodovico S. Staphylococcus aureus/Staphylococcus epidermidis from skin microbiota are balanced by Pomegranate peel extract: An eco-sustainable approach. PLoS One 2024; 19:e0308211. [PMID: 39088519 PMCID: PMC11293756 DOI: 10.1371/journal.pone.0308211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Accepted: 07/17/2024] [Indexed: 08/03/2024] Open
Abstract
The imbalance in skin microbiota is characterized by an increased number of pathogens in respect to commensal microorganisms. Starting from a skin microbiota collection, the aim of this work was to evaluate the possible role of Pomegranate (Punica granatum L.) Peel Extract (PPE) in restoring the skin microbiota balance acting on Staphylococcus spp. PPE was extracted following green methodology by using n-butane and the Dimethyl Ether (DME) solvents and analyzed for phytochemical composition and antimicrobial activity. The PPE antimicrobial action was evaluated against Gram +, Gram - bacteria and yeast reference strains and the most effective extract was tested against the main skin microbiota isolated strains. PPE extracted with DME showed the best antimicrobial action with MICs ranging from 1 to 128 mg/mL; the main active compounds were Catechin, Quercetin, Vanillic acid and Gallic acid. The PPE in DME anti-adhesive effect was examined against S. epidermidis and S. aureus mono and dual-species biofilm formation by biomass quantification and CFU/mL determination. The extract toxicity was evaluated by using Galleria mellonella larvae in vivo model. The extract displayed a significant anti-adhesive activity with a remarkable species-specific action at 4 and 8 mg/mL against S. epidermidis and S. aureus mono and dual-species biofilms. PPE in DME could represent an eco-sustainable non-toxic strategy to affect the Staphylococcal skin colonization in a species-specific way. The innovation of this work is represented by the reuse of food waste to balance skin microbiota.
Collapse
Affiliation(s)
- Sara D’Arcangelo
- Department of Pharmacy, University “G. d’Annunzio” Chieti-Pescara, Chieti, Italy
| | - Paola Di Fermo
- Department of Medical, Oral and Biotechnological Sciences, University “G. d’Annunzio” Chieti- Pescara, Chieti, Italy
| | - Firas Diban
- Department of Pharmacy, University “G. d’Annunzio” Chieti-Pescara, Chieti, Italy
| | - Vincenzo Ferrone
- Department of Pharmacy, University “G. d’Annunzio” Chieti-Pescara, Chieti, Italy
| | - Simonetta D’Ercole
- Department of Medical, Oral and Biotechnological Sciences, University “G. d’Annunzio” Chieti- Pescara, Chieti, Italy
| | - Mara Di Giulio
- Department of Pharmacy, University “G. d’Annunzio” Chieti-Pescara, Chieti, Italy
| | - Silvia Di Lodovico
- Department of Pharmacy, University “G. d’Annunzio” Chieti-Pescara, Chieti, Italy
| |
Collapse
|