1
|
Lopes GF, Oliveira VDC, Monteiro RM, Amadio Domingues PC, Bim FL, Bim LL, Ferreira da Silva GB, Pereira Dos Santos A, do Nascimento C, de Andrade D, Watanabe E. Assessment of peripheral venous catheters microbiota and its association with phlebitis. Infect Dis Health 2025; 30:1-11. [PMID: 39107205 DOI: 10.1016/j.idh.2024.07.005] [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: 12/11/2023] [Revised: 07/07/2024] [Accepted: 07/09/2024] [Indexed: 08/09/2024]
Abstract
BACKGROUND Peripheral venous catheters (PVCs) remain the primary mode of short-term venous access for managing intravenous fluid, obtaining blood samples, and peripheral parenteral nutrition. They may get contaminated and require regular monitoring to prevent complications. This study evaluated the occurrence of phlebitis and its associated-clinical and microbiological indicators. METHODS The frequency of phlebitis was evaluated in hospitalized patients of both medical and surgical fields. Subsequently, the dichotomous association between the presence of phlebitis and the clinical aspects was investigated. In parallel, the bacterial contamination of PVCs was assessed through culture-based methods, microscopy observation, and 16S rRNA gene sequencing. RESULTS Approximately one in four patients presented phlebitis (28.4%). The most frequent symptom was erythema at access site, with or without pain, corresponding to Score 1 on the phlebitis scale (17.9%). Colonization of both lumen and external surface of PVC was observed in 31.3% of the samples. Staphylococcus and Pseudomonas were the most isolated bacterial genera on the PVC surface. No significant association was observed between the presence of phlebitis and the clinical aspects, as well as the presence of microorganisms. CONCLUSION Microorganism were present on both internal and external PVC surface, without being associated to phlebitis.
Collapse
Affiliation(s)
- Gustavo Francisco Lopes
- Clinical Hospital of Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil; Human Exposome and Infectious Diseases Network (HEID), School of Nursing of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Viviane de Cássia Oliveira
- Human Exposome and Infectious Diseases Network (HEID), School of Nursing of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil; Department of Materials and Prostheses, School of Dentistry of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil.
| | - Rachel Maciel Monteiro
- Human Exposome and Infectious Diseases Network (HEID), School of Nursing of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Pedro Castania Amadio Domingues
- Human Exposome and Infectious Diseases Network (HEID), School of Nursing of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Felipe Lazarini Bim
- Human Exposome and Infectious Diseases Network (HEID), School of Nursing of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Lucas Lazarini Bim
- Human Exposome and Infectious Diseases Network (HEID), School of Nursing of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Gabriela Bassi Ferreira da Silva
- Human Exposome and Infectious Diseases Network (HEID), School of Nursing of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - André Pereira Dos Santos
- Human Exposome and Infectious Diseases Network (HEID), School of Nursing of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Cássio do Nascimento
- Department of Materials and Prostheses, School of Dentistry of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Denise de Andrade
- Human Exposome and Infectious Diseases Network (HEID), School of Nursing of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Evandro Watanabe
- Human Exposome and Infectious Diseases Network (HEID), School of Nursing of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil; Department of Restorative Dentistry, School of Dentistry of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| |
Collapse
|
2
|
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
|
3
|
Ren X, Zhou N, Li D, Li L, Wang Y, Li L, Ma Y, Gao X, Zhao Y, Sun Y, Wang Y. Network pharmacology, transcriptomics, and biological validation reveal a lipid secretion inhibitory and anti-inflammatory mechanism of tanreqing gel in the treatment of acne. JOURNAL OF ETHNOPHARMACOLOGY 2025; 340:119278. [PMID: 39719227 DOI: 10.1016/j.jep.2024.119278] [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: 09/18/2024] [Revised: 12/20/2024] [Accepted: 12/20/2024] [Indexed: 12/26/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Acne vulgaris is a common skin disease affecting the pilosebaceous unit, in which abnormal sebum secretion and inflammation play crucial roles. The traditional Chinese medicine Tanreqing has been utilized in dermatology to effectively treat various diseases. However, its effects and underlying mechanisms in acne vulgaris remain unclear. AIM OF THE STUDY This study aims to assess the potential benefits of Tanreqing gel (TRQ) in acne treatment and to explore the mechanisms by which TRQ inhibits sebum secretion and reduces inflammation. MATERIALS AND METHODS A mouse model of acne induced by Cutibacterium acnes (C. acnes) was established. The impact of TRQ on acne lesions was assessed using optical imaging and histopathology. Network pharmacology and transcriptomics were used to identify significant intervention pathways and targets. Both in vivo and in vitro experiments were conducted to detect the expression of genes and proteins associated with inflammation and sebum metabolism. RESULTS TRQ significantly improved pathological changes in the lesion areas of mice, such as redness, vascular dilation, and increased blood flow. It also reduced inflammatory cell infiltration in the dermis and inhibited the accumulation of lipids in the sebaceous glands. Network pharmacology analysis indicated that TRQ might exert anti-inflammatory effects through the IL-17, TOLL-like receptor, and NF-κB signaling pathways. The transcriptomic analysis confirmed the importance of these pathways in the C. acnes-induced acne model. Furthermore, TRQ was found to reduce sebum secretion by inhibiting fatty acid biosynthesis through the suppression of proteins in the PI3K-Akt signaling pathway. Cell experiments confirmed that TRQ could suppress the release of inflammatory factors induced by C. acnes surface structure peptidoglycan (PGN) and metabolite porphyrins. Additionally, it was observed to reverse the elevated porphyrin secretion associated with abnormal sebum production, ultimately relieving acne inflammation. CONCLUSION This study demonstrated that TRQ effectively alleviates C. acnes-induced acne symptoms by inhibiting sebum secretion and inflammatory responses through multiple pathways and targets. It provides new insights and directions for acne treatment.
Collapse
Affiliation(s)
- Xing Ren
- Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing, China
| | - Na Zhou
- Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing, China
| | - Dongying Li
- Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing, China
| | - Lu Li
- Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yunong Wang
- Beijing University of Chinese Medicine Third Affiliated Hospital, Beijing, China
| | - Lishuang Li
- Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yuman Ma
- Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xinyu Gao
- Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yujia Zhao
- Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yanan Sun
- Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing, China.
| | - Yi Wang
- Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing, China.
| |
Collapse
|
4
|
Lei Y, Li M, Zhang H, Deng Y, Dong X, Chen P, Li Y, Zhang S, Li C, Wang S, Tao R. Comparative analysis of the human microbiome from four different regions of China and machine learning-based geographical inference. mSphere 2025; 10:e0067224. [PMID: 39699186 PMCID: PMC11774049 DOI: 10.1128/msphere.00672-24] [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: 08/11/2024] [Accepted: 11/22/2024] [Indexed: 12/20/2024] Open
Abstract
The human microbiome, the community of microorganisms that reside on and inside the human body, is critically important for health and disease. However, it is influenced by various factors and may vary among individuals residing in distinct geographic regions. In this study, 220 samples, consisting of sterile swabs from palmar skin and oral and nasal cavities were collected from Chinese Han individuals living in Shanghai, Chifeng, Kunming, and Urumqi, representing the geographic regions of east, northeast, southwest, and northwest China. The full-length 16S rRNA gene of the microbiota in each sample was sequenced using the PacBio single-molecule real-time sequencing platform, followed by clustering the sequences into operational taxonomic units (OTUs). The analysis revealed significant differences in microbial communities among the four regions. Cutibacterium was the most abundant bacterium in palmar samples from Shanghai and Kunming, Psychrobacter in Chifeng samples, and Psychrobacillus in Urumqi samples. Additionally, Streptococcus and Staphylococcus were the dominant bacteria in the oral and nasal cavities. Individuals from the four regions could be distinguished and predicted based on a model constructed using the random forest algorithm, with the predictive effect of palmar microbiota being better than that of oral and nasal cavities. The prediction accuracy using hypervariable regions (V3-V4 and V4-V5) was comparable with that of using the entire 16S rRNA. Overall, our study highlights the distinctiveness of the human microbiome in individuals living in these four regions. Furthermore, the microbiome can serve as a biomarker for geographic origin inference, which has immense application value in forensic science.IMPORTANCEMicrobial communities in human hosts play a significant role in health and disease, varying in species, quantity, and composition due to factors such as gender, ethnicity, health status, lifestyle, and living environment. The characteristics of microbial composition at various body sites of individuals from different regions remain largely unexplored. This study utilized single-molecule real-time sequencing technology to detect the entire 16S rRNA gene of bacteria residing in the palmar skin, oral, and nasal cavities of Han individuals from four regions in China. The composition and structure of the bacteria at these three body sites were well characterized and found to differ regionally. The results elucidate the differences in bacterial communities colonizing these body sites across different regions and reveal the influence of geographical factors on human bacteria. These findings not only contribute to a deeper understanding of the diversity and geographical distribution of human bacteria but also enrich the microbiome data of the Asian population for further studies.
Collapse
Affiliation(s)
- Yinlei Lei
- Shanghai Key Laboratory of Forensic Medicine, Shanghai Forensic Service Platform, Academy of Forensic Sciences, Key Laboratory of Forensic Science, Ministry of Justice, Shanghai, China
- Department of Forensic Medicine, Zunyi Medical University, Zunyi, China
| | - Min Li
- School of Clinical and Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Han Zhang
- Institute of Forensic Science, Fudan University, Shanghai, China
- Department of Forensic Medicine, Guizhou Medical University, Guiyang, China
| | - Yu Deng
- Shanghai Key Laboratory of Forensic Medicine, Shanghai Forensic Service Platform, Academy of Forensic Sciences, Key Laboratory of Forensic Science, Ministry of Justice, Shanghai, China
- Department of Forensic Medicine, Zunyi Medical University, Zunyi, China
| | - Xinyu Dong
- Minhang Branch of Shanghai Public Security Bureau, Shanghai, China
| | - Pengyu Chen
- Department of Forensic Medicine, Zunyi Medical University, Zunyi, China
| | - Ye Li
- Department of Forensic Medicine, School of Basic Medical Sciences, Xinjiang Medical University, Urumqi, China
| | - Suhua Zhang
- Institute of Forensic Science, Fudan University, Shanghai, China
| | - Chengtao Li
- Institute of Forensic Science, Fudan University, Shanghai, China
| | - Shouyu Wang
- Department of Forensic Medicine, Shanghai Medical College, Fudan University, Shanghai, China
| | - Ruiyang Tao
- Shanghai Key Laboratory of Forensic Medicine, Shanghai Forensic Service Platform, Academy of Forensic Sciences, Key Laboratory of Forensic Science, Ministry of Justice, Shanghai, China
| |
Collapse
|
5
|
Escobar Marcillo DI, Privitera GF, Rollo F, Latini A, Giuliani E, Benevolo M, Giuliani M, Pichi B, Pellini R, Donà MG. Microbiome analysis in individuals with human papillomavirus oral infection. Sci Rep 2025; 15:2953. [PMID: 39848958 PMCID: PMC11757712 DOI: 10.1038/s41598-024-81607-4] [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: 02/23/2024] [Accepted: 11/27/2024] [Indexed: 01/25/2025] Open
Abstract
Microbiome gained attention as a cofactor in cancers originating from epithelial tissues. High-risk (hr)HPV infection causes oropharyngeal squamous cell carcinoma but only in a fraction of hrHPV+ individuals, suggesting that other factors play a role in cancer development. We investigated oral microbiome in cancer-free subjects harboring hrHPV oral infection (n = 33) and matched HPV- controls (n = 30). DNA purified from oral rinse-and-gargles of HIV-infected (HIV+) and HIV-uninfected (HIV-) individuals were used for 16S rRNA gene V3-V4 region amplification and sequencing. Analysis of differential microbial abundance and differential pathway abundance was performed, separately for HIV+ and HIV- individuals. Significant differences in alpha (Chao-1 and Shannon indices) and beta diversity (unweighted UniFrac distance) were observed between hrHPV+ and HPV-negative subjects, but only for the HIV- individuals. Infection by hrHPVs was associated with significant changes in the abundance of Saccharibacteria in HIV+ and Gracilibacteria in HIV- subjects. At the genus level, the greatest change in HIV+ individuals was observed for Bulleidia, which was significantly enriched in hrHPV+ subjects. In HIV- individuals, those hrHPV+ showed a significant enrichment of Parvimonas and depletion of Alloscardovia. Our data suggest a possible interplay between hrHPV infection and oral microbiome, which may vary with the HIV status.
Collapse
Affiliation(s)
- David Israel Escobar Marcillo
- Section of Mechanisms, Biomarkers and Models, Department of Environment and Health, Istituto Superiore di Sanità, Rome, Italy
| | - Grete Francesca Privitera
- Bioinformatics Unit, Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Francesca Rollo
- Pathology Department, IRCCS Regina Elena National Cancer Institute, Rome, Italy.
| | - Alessandra Latini
- STI/HIV Unit, San Gallicano Dermatological Institute IRCCS, Rome, Italy
| | - Eugenia Giuliani
- STI/HIV Unit, San Gallicano Dermatological Institute IRCCS, Rome, Italy
| | - Maria Benevolo
- Pathology Department, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Massimo Giuliani
- STI/HIV Unit, San Gallicano Dermatological Institute IRCCS, Rome, Italy
| | - Barbara Pichi
- Otolaryngology Head Neck Surgery Department, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Raul Pellini
- Otolaryngology Head Neck Surgery Department, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | | |
Collapse
|
6
|
Alperovitz CH, Ben David N, Ramot Y, Gross A, Mizrahi B. Living Microneedles for Intradermal Delivery of Beneficial Bacteria. ACS Biomater Sci Eng 2025. [PMID: 39828947 DOI: 10.1021/acsbiomaterials.4c02230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 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
|
7
|
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
|
8
|
Gu J, Wang J, Li Y, Li L, Zou Y, Guo Y, Yu B. Global burden of bacterial skin diseases from 1990 to 2045: an analysis based on global burden disease data. Arch Dermatol Res 2025; 317:266. [PMID: 39820797 DOI: 10.1007/s00403-025-03804-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 12/17/2024] [Accepted: 01/03/2025] [Indexed: 01/19/2025]
Abstract
Bacterial skin diseases are a category of inflammatory skin conditions caused by bacterial infections, which impose a significant global disease burden. However, they have not been well assessed or predicted on a global scale. It is necessary to update the estimates and forecast future trends of the global burden of bacterial skin diseases to evaluate the impact of past healthcare policies and to provide guidance and information for new national and international healthcare strategies. We aimed to describe the burden and trend of bacterial skin diseases and to predict the burden up to 2045. To achieve this, we employed a cross-sectional analysis based on Global Burden of Disease (GBD) data, utilizing advanced statistical models to quantify trends and forecast future burdens. Data on incidence and disability-adjusted life years (DALYs) of bacterial skin diseases were obtained from Global Burden of Disease 2021. We used average annual percent change (AAPC) by Joinpoint Regression to quantify the temporal trends. We conducted decomposition analysis to understand the contribution of aging, epidemiological changes, and population growth. Bayesian Age-Period-Cohort model was used to predict burden up to 2045. Global incidence rate of bacterial skin diseases increased from 8,988.74 per 100,000 in 1990 to 10,823.88 per 100,000, with AAPC of 0.62% (0.61 ~ 0.63%). The highest incidence rate was in low Socio-demographic Index (SDI) region and population aged < 35. The DALYs rate increased from 20.82 per 100,000 in 1990 to 25.45 per 100,000 in 2021, with AAPC of -0.11% (-0.34 ~ 0.13%). The highest increase of DALYs was in high SDI region and population aged > 85. Among the three evaluated factors of decomposition analysis, the major drivers of incident case rise were population growth, followed by epidemiological changes; the major drivers of DALYs case rise were population growth, followed by aging. The number of incidence cases has increased since 1990, reaching nearly 90 million in 2021 and expected to hit 1.2 billion in 2045. The incidence rate has also risen. Meanwhile, DALYs showed an upward trend from 1990 to 2005, peaking at 32/100,000, then a downward trend. Our findings align with our initial objectives, demonstrating a significant increase in the global incidence of bacterial skin diseases and highlighting the need for targeted prevention and treatment strategies. The variation in burden across different regions and age groups underscores the importance of tailored public health interventions. Predictive models suggest a continued rise in incidence rates and incident cases through 2045, emphasizing the urgency for action. This study provides a foundation for future research and policy development aimed at reducing the burden of bacterial skin diseases worldwide.
Collapse
Affiliation(s)
- Jiaxu Gu
- Department of Dermatology, Shenzhen Key Discipline of Dermatology, Shenzhen Key Laboratory for Translational Medicine of Dermatology, Biomedical Research Institute, Institute of Dermatology, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Peking University, Shenzhen, China
| | - Jiaming Wang
- Department of Dermatology, Shenzhen Key Discipline of Dermatology, Shenzhen Key Laboratory for Translational Medicine of Dermatology, Biomedical Research Institute, Institute of Dermatology, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shantou University Medical College, Shenzhen, China
| | - Yannan Li
- Faculty of Medicine, School of Population and Public Health, University of British Columbia, Vancouver, Canada
| | - Lianjie Li
- Warren Skin Care Center, Phillipsburg, USA
| | - Yanfen Zou
- Department of Dermatology, Shenzhen Key Discipline of Dermatology, Shenzhen Key Laboratory for Translational Medicine of Dermatology, Biomedical Research Institute, Institute of Dermatology, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, China
| | - Yang Guo
- Department of Epidemiology and Statistics, School of Public Health, Hebei Key Laboratory of Environment and Human Health, Hebei Medical University, Shijiazhuang, Hebei Province, 050017, China.
| | - Bo Yu
- Department of Dermatology, Shenzhen Key Discipline of Dermatology, Shenzhen Key Laboratory for Translational Medicine of Dermatology, Biomedical Research Institute, Institute of Dermatology, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, China.
- Department of Dermatology, Peking University Shenzhen Hospital, Shenzhen, Guangdong Province, 518036, China.
| |
Collapse
|
9
|
Govender P, Ghai M. Population-specific differences in the human microbiome: Factors defining the diversity. Gene 2025; 933:148923. [PMID: 39244168 DOI: 10.1016/j.gene.2024.148923] [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: 05/24/2024] [Revised: 08/15/2024] [Accepted: 09/03/2024] [Indexed: 09/09/2024]
Abstract
Differences in microbial communities at different body habitats define the microbiome composition of the human body. The gut, oral, skin vaginal fluid and tissue microbiome, are pivotal for human development and immune response and cross talk between these microbiomes is evident. Population studies reveal that various factors, such as host genetics, diet, lifestyle, aging, and geographical location are strongly associated with population-specific microbiome differences. The present review discusses the factors that shape microbiome diversity in humans, and microbiome differences in African, Asian and Caucasian populations. Gut microbiome studies show that microbial species Bacteroides is commonly found in individuals living in Western countries (Caucasian populations), while Prevotella is prevalent in non-Western countries (African and Asian populations). This association is mainly due to the high carbohydrate, high fat diet in western countries in contrast to high fibre, low fat diets in African/ Asian regions. Majority of the microbiome studies focus on the bacteriome component; however, interesting findings reveal that increased bacteriophage richness, which makes up the virome component, correlates with decreased bacterial diversity, and causes microbiome dysbiosis. An increase of Caudovirales (bacteriophages) is associated with a decrease in enteric bacteria in inflammatory bowel diseases. Future microbiome studies should evaluate the interrelation between bacteriome and virome to fully understand their significance in the pathogenesis and progression of human diseases. With ethnic health disparities becoming increasingly apparent, studies need to emphasize on the association of population-specific microbiome differences and human diseases, to develop microbiome-based therapeutics. Additionally, targeted phage therapy is emerging as an attractive alternative to antibiotics for bacterial infections. With rapid rise in microbiome research, focus should be on standardizing protocols, advanced bioinformatics tools, and reducing sequencing platform related biases. Ultimately, integration of multi-omics data (genomics, transcriptomics, proteomics and metabolomics) will lead to precision models for personalized microbiome therapeutics advancement.
Collapse
Affiliation(s)
- Priyanka Govender
- Discipline of Genetics, School of Life Sciences, University of KwaZulu-Natal, Westville, South Africa
| | - Meenu Ghai
- Discipline of Genetics, School of Life Sciences, University of KwaZulu-Natal, Westville, South Africa.
| |
Collapse
|
10
|
Bayer IS. Fungal quorum sensing molecules as potential drugs in the treatment of chronic wounds and their delivery. Expert Opin Drug Deliv 2025:1-20. [PMID: 39791701 DOI: 10.1080/17425247.2025.2452303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 11/27/2024] [Accepted: 01/08/2025] [Indexed: 01/12/2025]
Abstract
INTRODUCTION Chronic non-healing wounds have emerged as a significant global healthcare challenge. Biofilm induced wound infections has been widely acknowledged. Despite the advanced understanding of biofilm formation, the existing approaches for diagnosing biofilms in wounds remain considerably suboptimal. Chemical signals produced by fungi to sense their environment, known as quorum sensing (QS) molecules are anticipated to cause revolution in non-healing wound antisepsis. AREAS COVERED Biofilms render chronic wounds resistant to treatment and impede tissue repair by inducing chronic inflammation. QS is a biochemical signaling pathway that involves certain secreted molecules, namely phenylethanoids, indolyl, and sesquiterpene alcohols that can significantly minimize and obliterate bacterial biofilms if properly applied and released in wound treatments. EXPERT OPINION QS molecules (QSMs) possess inhibitory properties that obstruct the formation of microbial biofilms and exhibit synergism with common antimicrobials. They can disrupt biofilms formed by drug-resistant microorganisms. The understanding of the current mechanisms and advancements in the utilization of QSMs within diverse drug delivery systems, and their release dynamics will be crucial in new drug design and delivery. Exploration of co-delivery of drugs alongside QS molecules, and assessing their impact on healing of chronic wounds before moving to clinical trials remain unaddressed.
Collapse
Affiliation(s)
- Ilker S Bayer
- Smart Materials, Istituto Italiano di Tecnologia, Genova, Italy
| |
Collapse
|
11
|
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
|
12
|
Pagac MP, Davient B, Plado LA, Lam HYI, Lee SM, Ravikrishnan A, Chua WLE, Muralidharan S, Sridharan A, Irudayaswamy AS, Srinivas R, Wearne S, Mohamed Naim AN, Ho EXP, Ng HQA, Kwah JS, Png E, Bendt AK, Wenk MR, Torta F, Nagarajan N, Common J, Chong YS, Tham EH, Shek LPC, Loo EXL, Chambers J, Yew YW, Loh M, Dawson TL. Life stage impact on the human skin ecosystem: lipids and the microbial community. NPJ Biofilms Microbiomes 2025; 11:13. [PMID: 39800795 PMCID: PMC11725588 DOI: 10.1038/s41522-025-00652-7] [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: 02/22/2024] [Accepted: 01/06/2025] [Indexed: 01/16/2025] Open
Abstract
Sebaceous free fatty acids are metabolized by multiple skin microbes into bioactive lipid mediators termed oxylipins. This study investigated correlations between skin oxylipins and microbes on the superficial skin of pre-pubescent children (N = 36) and adults (N = 100), including pre- (N = 25) and post-menopausal females (N = 25). Lipidomics and metagenomics revealed that Malassezia restricta positively correlated with the oxylipin 9,10-DiHOME on adult skin and negatively correlated with its precursor, 9,10-EpOME, on pre-pubescent skin. Co-culturing Malassezia with keratinocytes demonstrated a link between 9,10-DiHOME and pro-inflammatory cytokines IL-1β and IL-6 production. We also observed strong correlations between other skin oxylipins and microbial taxa, highlighting life stage differences in sebum production and microbial community composition. Our findings imply a complex host-microbe communication system mediated by lipid metabolism occurring on human skin, warranting further research into its role in skin health and disease and paving the way towards novel therapeutic targets and treatments.
Collapse
Grants
- This research was supported by the Singapore National Research Foundation under its Translational and Clinical Research (TCR) Flagship Programme and administered by the Singapore Ministry of Health’s National Medical Research Council (NMRC), Singapore - NMRC/TCR/004-NUS/2008; NMRC/TCR/012-NUHS/2014. Additional funding was provided by the Singapore Institute for Clinical Sciences, Agency for Science Technology and Research (A*STAR), Singapore. This study was also supported by Agency for Science, Technology and Research (A*STAR) BMRC EDB IAF-PP grant (H17/01/a0/004) (TD); Skin Research Institute of Singapore, IAF-PP (HBMS) grant; Asian Skin Microbiome Program IAF-PP grants (H18/01/a0/016) (TD) and (H22/J1/a0/040). HELIOS study (NTU IRB: 2016-11-030) is supported by the Singapore Ministry of Health’s National Medical Research Council under its OF-LCG funding scheme (NMRC Project Ref. MOH-000271-00), STaR funding scheme (NMRC Project Ref. NMRC/STaR/0028/2017) and intramural funding from Nanyang Technological University, Lee Kong Chian School of Medicine and the National Healthcare Group. The HELIOS study team is also supported by a team of outstanding operational and administrative staff.
- "This research was supported by the Singapore National Research Foundation under its Translational and Clinical Research (TCR) Flagship Programme and administered by the Singapore Ministry of Health’s National Medical Research Council (NMRC), Singapore - NMRC/TCR/004-NUS/2008; NMRC/TCR/012-NUHS/2014. Additional funding was provided by the Singapore Institute for Clinical Sciences, Agency for Science Technology and Research (A*STAR), Singapore. This study was also supported by Agency for Science, Technology and Research (A*STAR) BMRC EDB IAF-PP grant (H17/01/a0/004) (TD); Skin Research Institute of Singapore, IAF-PP (HBMS) grant; Asian Skin Microbiome Program IAF-PP grants (H18/01/a0/016) (TD) and (H22/J1/a0/040). HELIOS study (NTU IRB: 2016-11-030) is supported by the Singapore Ministry of Health’s National Medical Research Council under its OF-LCG funding scheme (NMRC Project Ref. MOH-000271-00), STaR funding scheme (NMRC Project Ref. NMRC/STaR/0028/2017) and intramural funding from Nanyang Technological University, Lee Kong Chian School of Medicine and the National Healthcare Group. The HELIOS study team is also supported by a team of outstanding operational and administrative staff."
Collapse
Affiliation(s)
- Martin P Pagac
- A*STAR Skin Research Labs (A*SRL), Agency for Science, Technology, and Research (A*STAR) & Skin Research Institute of Singapore (SRIS), Singapore, Republic of Singapore
- DSM-Firmenich, Perfumery and Beauty, Wurmisweg 576, Kaiseraugst, Switzerland
| | - Bala Davient
- A*STAR Skin Research Labs (A*SRL), Agency for Science, Technology, and Research (A*STAR) & Skin Research Institute of Singapore (SRIS), Singapore, Republic of Singapore
| | - Luca Antonio Plado
- A*STAR Skin Research Labs (A*SRL), Agency for Science, Technology, and Research (A*STAR) & Skin Research Institute of Singapore (SRIS), Singapore, Republic of Singapore
- Laboratory for Lipidomics and Lipid Biology, University of Manchester, Division of Pharmacy and Optometry, Faculty of Biology Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, Manchester, UK
| | - Hilbert Yuen In Lam
- A*STAR Skin Research Labs (A*SRL), Agency for Science, Technology, and Research (A*STAR) & Skin Research Institute of Singapore (SRIS), Singapore, Republic of Singapore
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Dr, Singapore, Singapore
| | - Shi Mun Lee
- A*STAR Skin Research Labs (A*SRL), Agency for Science, Technology, and Research (A*STAR) & Skin Research Institute of Singapore (SRIS), Singapore, Republic of Singapore
| | - Aarthi Ravikrishnan
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A*STAR), 60 Biopolis Street, Genome, Singapore, Republic of Singapore
| | - Wee Ling Esther Chua
- SLING, Singapore Lipidomics Incubator, Life Sciences Institute, National University of Singapore, Singapore, Singapore
| | - Sneha Muralidharan
- SLING, Singapore Lipidomics Incubator, Life Sciences Institute, National University of Singapore, Singapore, Singapore
| | - Aishwarya Sridharan
- A*STAR Skin Research Labs (A*SRL), Agency for Science, Technology, and Research (A*STAR) & Skin Research Institute of Singapore (SRIS), Singapore, Republic of Singapore
| | - Antony S Irudayaswamy
- A*STAR Skin Research Labs (A*SRL), Agency for Science, Technology, and Research (A*STAR) & Skin Research Institute of Singapore (SRIS), Singapore, Republic of Singapore
| | - Ramasamy Srinivas
- A*STAR Skin Research Labs (A*SRL), Agency for Science, Technology, and Research (A*STAR) & Skin Research Institute of Singapore (SRIS), Singapore, Republic of Singapore
- Cellivate Technologies, Singapore, Singapore
| | - Stephen Wearne
- A*STAR Skin Research Labs (A*SRL), Agency for Science, Technology, and Research (A*STAR) & Skin Research Institute of Singapore (SRIS), Singapore, Republic of Singapore
| | - Ahmad Nazri Mohamed Naim
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A*STAR), 60 Biopolis Street, Genome, Singapore, Republic of Singapore
| | - Eliza Xin Pei Ho
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A*STAR), 60 Biopolis Street, Genome, Singapore, Republic of Singapore
| | - H Q Amanda Ng
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A*STAR), 60 Biopolis Street, Genome, Singapore, Republic of Singapore
| | - Junmei Samantha Kwah
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A*STAR), 60 Biopolis Street, Genome, Singapore, Republic of Singapore
| | - Eileen Png
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A*STAR), 60 Biopolis Street, Genome, Singapore, Republic of Singapore
| | - Anne K Bendt
- SLING, Singapore Lipidomics Incubator, Life Sciences Institute, National University of Singapore, Singapore, Singapore
| | - Markus R Wenk
- SLING, Singapore Lipidomics Incubator, Life Sciences Institute, National University of Singapore, Singapore, Singapore
- Precision Medicine Translational Research Programme and Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Federico Torta
- SLING, Singapore Lipidomics Incubator, Life Sciences Institute, National University of Singapore, Singapore, Singapore
- Precision Medicine Translational Research Programme and Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Niranjan Nagarajan
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A*STAR), 60 Biopolis Street, Genome, Singapore, Republic of Singapore
| | - John Common
- A*STAR Skin Research Labs (A*SRL), Agency for Science, Technology, and Research (A*STAR) & Skin Research Institute of Singapore (SRIS), Singapore, Republic of Singapore
| | - Yap Seng Chong
- Institute for Human Development and Potential (IHDP), Agency for Science, Technology and Research (A*STAR), 30 Medical Drive, Singapore, Republic of Singapore
| | - Elizabeth Huiwen Tham
- Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore (NUS), Singapore, Singapore
- Khoo Teck Puat-National University Children's Medical Institute, National University Health System (NUHS), Singapore, Singapore
- Human Potential Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Lynette Pei-Chi Shek
- Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore (NUS), Singapore, Singapore
- Khoo Teck Puat-National University Children's Medical Institute, National University Health System (NUHS), Singapore, Singapore
| | - Evelyn Xiu Ling Loo
- Institute for Human Development and Potential (IHDP), Agency for Science, Technology and Research (A*STAR), 30 Medical Drive, Singapore, Republic of Singapore
- Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore (NUS), Singapore, Singapore
- Human Potential Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - John Chambers
- Lee Kong Chian School of Medicine, Nanyang Technological University, Clinical Sciences Building, Singapore, Singapore
| | - Yik Weng Yew
- Lee Kong Chian School of Medicine, Nanyang Technological University, Clinical Sciences Building, Singapore, Singapore
| | - Marie Loh
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A*STAR), 60 Biopolis Street, Genome, Singapore, Republic of Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University, Clinical Sciences Building, Singapore, Singapore
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, St Mary's Campus, London, UK
- National Skin Centre, Singapore, Singapore
| | - Thomas L Dawson
- A*STAR Skin Research Labs (A*SRL), Agency for Science, Technology, and Research (A*STAR) & Skin Research Institute of Singapore (SRIS), Singapore, Republic of Singapore.
- Department of Drug Discovery, College of Pharmacy, Medical University of South Carolina, Charleston, USA.
| |
Collapse
|
13
|
Vanić Ž, Jøraholmen MW, Škalko-Basnet N. Challenges and considerations in liposomal hydrogels for the treatment of infection. Expert Opin Drug Deliv 2025:1-22. [PMID: 39797393 DOI: 10.1080/17425247.2025.2451620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Accepted: 01/07/2025] [Indexed: 01/13/2025]
Abstract
INTRODUCTION Liposomal hydrogels are novel drug delivery systems that comprise preformed liposomes incorporated in hydrogels destined for mostly localized drug therapy, herewith antimicrobial therapy. The formulation benefits from versatility of liposomes as lipid-based nanocarriers that enable delivery of various antimicrobials of different lipophilicities, and secondary vehicle, hydrogel, that assures better retention time of formulation at the infection site. Especially in an era of alarming antimicrobial resistance, efficient localized antimicrobial therapy that avoids systemic exposure of antimicrobial and related side effects is crucial. AREAS COVERED We provide an overview of liposomal hydrogels that were developed for superior delivery of antimicrobials at different infections sites, with focus on skin and vaginal infections. The review summarizes the challenges of infection site and most common infection-causing pathogens and offers commentary on most relevant features the formulation needs to optimize to increase the therapy outcome. We discuss the impact of liposomal composition, size, and choice of polymer-forming hydrogel on antimicrobial outcome based on the literature overview and own experience in the field. EXPERT OPINION Liposomal hydrogels offer improved therapy outcome in localized antimicrobial therapy. By fine-tuning of liposomal as well as hydrogel properties, formulations with superior performance can be optimized targeting specific infection site.
Collapse
Affiliation(s)
- Željka Vanić
- Department of Pharmaceutical Technology, Faculty of Pharmacy and Biochemistry, University of Zagreb, Zagreb, Croatia
| | - May Wenche Jøraholmen
- Drug Transport and Delivery Research Group, Department of Pharmacy, Faculty of Health Sciences, University of Tromsø The Arctic University of Norway, Tromsø, Norway
| | - Nataša Škalko-Basnet
- Drug Transport and Delivery Research Group, Department of Pharmacy, Faculty of Health Sciences, University of Tromsø The Arctic University of Norway, Tromsø, Norway
| |
Collapse
|
14
|
Heckmann ND, Culler M, Mont MA, Lieberman JR, Parvizi J. Emerging Concepts in Periprosthetic Joint Infection Research: The Human Microbiome. J Arthroplasty 2025:S0883-5403(25)00001-4. [PMID: 39798621 DOI: 10.1016/j.arth.2025.01.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Revised: 11/26/2024] [Accepted: 01/06/2025] [Indexed: 01/15/2025] Open
Abstract
Microorganisms, including bacteria, fungi, and viruses, that reside on and within the human body are collectively known as the human microbiome. Dysbiosis, or disruption in the microbiome, has been implicated in several disease processes, including asthma, obesity, autoimmune diseases, and numerous other conditions. While the Human Microbiome Project (HMP) and the generation of descriptive studies it inspired established correlations between characteristic patterns in the composition of the microbiome and specific disease phenotypes, current research has begun to focus on elucidating the causal role of the microbiome in disease pathogenesis. Within the field of orthopaedic surgery, researchers have proposed the concept of a "gut-joint axis" by which the intestinal microbiome influences joint health and the development of diseases such as osteoarthritis and periprosthetic joint infection (PJI). It is theorized that intestinal dysbiosis increases gut permeability, leading to the translocation of bacteria and their metabolic products into the systemic circulation and the stimulation of proinflammatory response cascades throughout the body, including within the joints. While correlative studies have identified patterns of dysbiotic derangement associated with osteoarthritis and PJI, translational research is needed to clarify the precise mechanisms by which these changes influence disease processes. Additionally, an emerging body of literature has challenged the previously held belief that certain body sites are sterile and do not possess a microbiome, with studies identifying distinct microbial genomic signatures and a core microbiome that varies between anatomic sites. A more thorough characterization of the joint microbiome may have profound implications for our understanding of PJI pathogenesis and our ability to stratify patients based on risk. The purpose of this review was to outline our current understanding of the human microbiome, to describe the gut-joint axis and its role in specific pathologies, including PJI, and to highlight the potential of microbiome-based therapeutic interventions in the field of orthopaedics.
Collapse
Affiliation(s)
- Nathanael D Heckmann
- Department of Orthopaedic Surgery, Keck School of Medicine of the University of Southern California, Los Angeles, CA, United States.
| | - McKenzie Culler
- Department of Orthopaedic Surgery, Keck School of Medicine of the University of Southern California, Los Angeles, CA, United States
| | - Michael A Mont
- LifeBridge Health, Sinai Hospital of Baltimore, The Rubin Institute for Advanced Orthopaedics, Baltimore, MD, United States
| | - Jay R Lieberman
- Department of Orthopaedic Surgery, Keck School of Medicine of the University of Southern California, Los Angeles, CA, United States
| | - Javad Parvizi
- International Joint Center, Acibadem University Hospital, Istanbul, Turkey
| |
Collapse
|
15
|
Xu Y, Wang Z, Li C, Tian S, Du W. Droplet microfluidics: unveiling the hidden complexity of the human microbiome. LAB ON A CHIP 2025. [PMID: 39775305 DOI: 10.1039/d4lc00877d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2025]
Abstract
The human body harbors diverse microbial communities essential for maintaining health and influencing disease processes. Droplet microfluidics, a precise and high-throughput platform for manipulating microscale droplets, has become vital in advancing microbiome research. This review introduces the foundational principles of droplet microfluidics, its operational capabilities, and wide-ranging applications. We emphasize its role in enhancing single-cell sequencing technologies, particularly genome and RNA sequencing, transforming our understanding of microbial diversity, gene expression, and community dynamics. We explore its critical function in isolating and cultivating traditionally unculturable microbes and investigating microbial activity and interactions, facilitating deeper insight into community behavior and metabolic functions. Lastly, we highlight its broader applications in microbial analysis and its potential to revolutionize human health research by driving innovations in diagnostics, therapeutic development, and personalized medicine. This review provides a comprehensive overview of droplet microfluidics' impact on microbiome research, underscoring its potential to transform our understanding of microbial dynamics and their relevance to health and disease.
Collapse
Affiliation(s)
- Yibin Xu
- State Key Laboratory of Microbial Resources, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China.
| | - Zhiyi Wang
- State Key Laboratory of Microbial Resources, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China.
- Medical School and College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Caiming Li
- State Key Laboratory of Microbial Resources, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China.
- Medical School and College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Shuiquan Tian
- State Key Laboratory of Microbial Resources, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China.
| | - Wenbin Du
- State Key Laboratory of Microbial Resources, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China.
- Medical School and College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
16
|
Szabó K, Balogh F, Romhányi D, Erdei L, Toldi B, Gyulai R, Kemény L, Groma G. Epigenetic Regulatory Processes Involved in the Establishment and Maintenance of Skin Homeostasis-The Role of Microbiota. Int J Mol Sci 2025; 26:438. [PMID: 39859154 PMCID: PMC11764776 DOI: 10.3390/ijms26020438] [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: 11/29/2024] [Revised: 12/30/2024] [Accepted: 01/02/2025] [Indexed: 01/27/2025] Open
Abstract
Epigenetic mechanisms are central to the regulation of all biological processes. This manuscript reviews the current understanding of diverse epigenetic modifications and their role in the establishment and maintenance of normal skin functions. In healthy skin, these mechanisms allow for the precise control of gene expression, facilitating the dynamic balance between cell proliferation and differentiation necessary for effective barrier function. Furthermore, as the skin ages, alterations in epigenetic marks can lead to impaired regenerative capacity and increased susceptibility to environmental stressors. The interaction between skin microbiota and epigenetic regulation will also be explored, highlighting how microbial communities can influence skin health by modulating the host gene expression. Future research should focus on the development of targeted interventions to promote skin development, resilience, and longevity, even in an ever-changing environment. This underscores the need for integrative approaches to study these complex regulatory networks.
Collapse
Affiliation(s)
- Kornélia Szabó
- HUN-REN-SZTE Dermatological Research Group, 6720 Szeged, Hungary (L.K.); (G.G.)
- Department of Dermatology and Allergology, Albert Szent-Györgyi Medical School, University of Szeged, 6720 Szeged, Hungary
- HCEMM-USZ Skin Research Group, 6720 Szeged, Hungary
| | - Fanni Balogh
- HUN-REN-SZTE Dermatological Research Group, 6720 Szeged, Hungary (L.K.); (G.G.)
| | - Dóra Romhányi
- Department of Dermatology and Allergology, Albert Szent-Györgyi Medical School, University of Szeged, 6720 Szeged, Hungary
| | - Lilla Erdei
- HUN-REN-SZTE Dermatological Research Group, 6720 Szeged, Hungary (L.K.); (G.G.)
- Department of Dermatology and Allergology, Albert Szent-Györgyi Medical School, University of Szeged, 6720 Szeged, Hungary
- HCEMM-USZ Skin Research Group, 6720 Szeged, Hungary
| | - Blanka Toldi
- HUN-REN-SZTE Dermatological Research Group, 6720 Szeged, Hungary (L.K.); (G.G.)
| | - Rolland Gyulai
- Department of Dermatology and Allergology, Albert Szent-Györgyi Medical School, University of Szeged, 6720 Szeged, Hungary
| | - Lajos Kemény
- HUN-REN-SZTE Dermatological Research Group, 6720 Szeged, Hungary (L.K.); (G.G.)
- Department of Dermatology and Allergology, Albert Szent-Györgyi Medical School, University of Szeged, 6720 Szeged, Hungary
- HCEMM-USZ Skin Research Group, 6720 Szeged, Hungary
| | - Gergely Groma
- HUN-REN-SZTE Dermatological Research Group, 6720 Szeged, Hungary (L.K.); (G.G.)
- Department of Dermatology and Allergology, Albert Szent-Györgyi Medical School, University of Szeged, 6720 Szeged, Hungary
| |
Collapse
|
17
|
Evans SE, Valentine ME, Gallimore F, Meka Y, Koehler SI, Yu HD, Valentovic MA, Long TE. Perturbations in the gut microbiome of C57BL/6 mice by the sobriety aid Antabuse® (disulfiram). J Appl Microbiol 2025; 136:lxae305. [PMID: 39701818 PMCID: PMC11704607 DOI: 10.1093/jambio/lxae305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 12/03/2024] [Accepted: 12/17/2024] [Indexed: 12/21/2024]
Abstract
AIMS Disulfiram (Antabuse®) is an oral alcohol sobriety medication that exhibits antimicrobial activity against Gram-positive facultative anaerobes. The aims of this study were to measure the antimicrobial activity against anaerobic bacteria of the gut human microbiome and establish the extent that disulfiram alters the microbial composition of the ileum, cecum, and feces using C57BL/6 mice. METHODS AND RESULTS Antimicrobial susceptibility testing by the microdilution method revealed that disulfiram inhibits the in vitro growth of gut anaerobic species of Bacteroides, Clostridium, Peptostreptococcus, and Porphyromonas. Differential sequencing of 16S rRNA isolated from the ileum, cecum, and feces contents of treated vs. untreated mice showed that disulfiram enriches the Gram-negative enteric population. In female mice, the enrichment was greatest in the ileum, whereas the feces composition in male mice was the most heavily altered. CONCLUSIONS Daily administration of oral disulfiram depletes the enteric Gram-positive anaerobe population as predicted by the minimum inhibitory concentration data for isolates from the human gut microbiota.
Collapse
Affiliation(s)
- Sarah E Evans
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25755, United States
| | - Meagan E Valentine
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25755, United States
| | - Fallon Gallimore
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25755, United States
| | - Yogesh Meka
- Department of Pharmaceutical Sciences, School of Pharmacy, Marshall University, Huntington, WV 25755, United States
| | - Samuel I Koehler
- Bioscience Division, Los Alamos National Laboratory, Los Alamos, NM 87545, United States
| | - Hongwei D Yu
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25755, United States
| | - Monica A Valentovic
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25755, United States
| | - Timothy E Long
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV 25755, United States
- Department of Pharmaceutical Sciences, School of Pharmacy, Marshall University, Huntington, WV 25755, United States
| |
Collapse
|
18
|
Hou K, Tan Y, Zhang Q. Investigating the causal relationship between skin microbiota and hypertrophic scar using bidirectional mendelian randomization. Burns 2025; 51:107376. [PMID: 39778466 DOI: 10.1016/j.burns.2025.107376] [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: 08/30/2024] [Revised: 12/16/2024] [Accepted: 01/04/2025] [Indexed: 01/11/2025]
Abstract
BACKGROUND Hypertrophic scar (HS) is acknowledged as a pathological fibro-proliferative disease of the dermis, resulting from excessive connective tissue growth. HS significantly impacts patient quality of life due to both social and functional issues. Despite various treatments, therapeutic effectiveness remains limited, necessitating further exploration of underlying factors and mechanisms. OBJECTIVE The current study was designed to determine the causal relationship between skin microbiota and HS employing a bidirectional Mendelian randomization (MR) approach. METHODS We utilized genome-wide association study (GWAS) data from the PopGen cohort and the FinnGen database. Independent single nucleotide polymorphisms (SNPs) linked to the skin microbiota were identified as instrumental variables (IVs) chosen for the two-sample MR analysis. Key analytical approaches included inverse variance weighting (IVW), MR-Egger, simple median, simple mode, and weighted mode, with MR-Egger intercept test and Cochrane's Q test used to detect potential horizontal pleiotropy and heterogeneity. RESULTS The two-sample MR analysis identified significant causal relationships between specific skin microbiota features and HS. Notably, Enhydrobacter, Micrococcus, and Acinetobacter on moist skin exhibited protective effects against HS, whereas Finegoldia and Lactobacillales on dry skin were linked to an increased risk of HS. Sensitivity analyses verified the strength of these results, revealing no notable horizontal pleiotropy or heterogeneity. CONCLUSION Our research reveals a unidirectional causal relationship between certain skin microbiota and HS, suggesting that modulation of skin microbiota could be a novel therapeutic approach for HS management. These results emphasize the significance of considering skin microbiota in the pathogenesis and treatment of HS.
Collapse
Affiliation(s)
- Kai Hou
- Department of Plastic and Cosmetic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yufang Tan
- Department of Plastic and Cosmetic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Qi Zhang
- Department of Plastic and Cosmetic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| |
Collapse
|
19
|
Calvez A, Stien D, Villette C, Lebaron P, Ravard Helffer K, Doat G, Bourrain M. The association of dextran sodium sulfate to the bioactive agent I-modulia® attenuates Staphylococcus aureus virulence expression and δ-toxin production. Microb Pathog 2025; 198:107163. [PMID: 39608513 DOI: 10.1016/j.micpath.2024.107163] [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: 09/16/2024] [Revised: 11/22/2024] [Accepted: 11/25/2024] [Indexed: 11/30/2024]
Abstract
As a part of the human skin commensal bacterial community, Staphylococcus aureus contributes to the host's immune system education. Nevertheless, it is also considered as an opportunistic pathogen involved in cutaneous infections or skin pathologies, in particular atopic dermatitis. To switch to a pathogenic behavior, S. aureus uses regulatory mechanisms to collectively produce virulence factors. Deprivation of these factors has emerged as a promising way to prevent or treat Staphylococcal diseases in facilitating the role of the immune system, while preserving the protective one of the commensal communities. This study focuses on the anti-virulent effect of dextran sodium sulfate (DSS) and I-modulia®, two natural products that have already proven their value in skincare. The anti-virulent capacity of DSS was first demonstrated by a dose-dependent inhibition of δ-toxin release, a virulence factor known to be a potent inducer of mast cell degranulation, on in vitro S. aureus cultures at high and low virulent states. A transcriptomic study was then implemented for a comprehensive overview of the anti-virulent impact. The results have shown the downregulation of many transcripts related to host immune evasion (scn, sbi), as well as exotoxins (α,γ-toxin) and adhesins production (map, emp), mostly under the control of SaeRS Two-Component System (TCS), one of the two major virulence regulators in S. aureus. Interestingly, genes related to secretion systems and the synthesis of exo-proteases were significantly downregulated when DSS was used in combination with I-modulia®. The repression of these genes was not previously observed and reflects a broader inhibitory action. We have also demonstrated that the inhibition of virulence factors didn't affect S. aureus viability. Our findings suggest that combining DSS and I-modulia® could be a promising therapeutic strategy to counteract microbial dysbiosis in the treatment of S. aureus skin pathologies in re-empowering the host's natural immune defenses.
Collapse
Affiliation(s)
- A Calvez
- Pierre Fabre Dermo-Cosmétique et Personal Care, 31000, Toulouse, France
| | - D Stien
- Sorbonne Université, CNRS, Laboratoire de Biodiversité et Biotechnologies Microbiennes, LBBM, Observatoire Océanologique, 66650, Banyuls-sur-mer, France
| | - C Villette
- Pierre Fabre Dermo-Cosmétique et Personal Care, 31000, Toulouse, France
| | - P Lebaron
- Sorbonne Université, CNRS, Laboratoire de Biodiversité et Biotechnologies Microbiennes, LBBM, Observatoire Océanologique, 66650, Banyuls-sur-mer, France
| | - K Ravard Helffer
- Pierre Fabre Dermo-Cosmétique et Personal Care, 31000, Toulouse, France
| | - G Doat
- Direction Médicale AVENE, Lavaur, France
| | - M Bourrain
- Pierre Fabre Dermo-Cosmétique et Personal Care, 31000, Toulouse, France.
| |
Collapse
|
20
|
du Crest D, Wustrow P, Worsley O, Geusens B, Badri O, Madhumita M, Papier A, Zink A, Hædersdal M, Garibyan L. Skin & Digital: The 2023 Startups/Innovators. JID INNOVATIONS 2025; 5:100316. [PMID: 39493507 PMCID: PMC11530584 DOI: 10.1016/j.xjidi.2024.100316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2024] Open
Affiliation(s)
| | | | | | | | | | - Monisha Madhumita
- Department of Dermatology, Dermanalytica, Bangalore, India
- Department of Dermatology, Saveetha Medical College, SIMATS, Chennai, Tamilnadu, India
| | - Art Papier
- Department of Dermatology, University of Rochester College of Medicine, Rochester, New York, USA
| | - Alexander Zink
- Department of Dermatology and Allergy, School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Merete Hædersdal
- Department of Dermatology, Copenhagen University Hospital, Copenhagen, Denmark
| | - Lilit Garibyan
- Wellman Center for Photomedicine, Department of Dermatology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
21
|
Johansson A, Ho NPY, Takizawa H. Microbiome and Hemato-immune Aging. Exp Hematol 2025; 141:104685. [PMID: 39581302 DOI: 10.1016/j.exphem.2024.104685] [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: 09/08/2024] [Revised: 10/17/2024] [Accepted: 11/16/2024] [Indexed: 11/26/2024]
Abstract
The microbiome is a highly complex and diverse symbiotic component that undergoes dynamic changes with the organismal aging. Microbial perturbations, termed dysbiosis, exert strong influence on dysregulating the bone marrow niche and subsequently promoting the aging of hematopoietic and immune system. Accumulating studies have revealed the substantial impact of intestinal microbiome on the initiation and progression of age-related hematologic alteration and diseases, such as clonal hematopoiesis and blood cancers. Current therapeutic approaches to restore the altered microbiome diversity target specific pathobionts and are demonstrated to improve clinical outcomes of antihematologic malignancy treatments. In this review, we discuss the interplay between the microbiome and the hemato-immune system during aging process. We also shed light on the emerging therapeutic strategies to tackle the dysbiosis for amelioration of aging and disease progression.
Collapse
Affiliation(s)
- Alban Johansson
- Laboratory of Stem Cell Stress, International Research Center for Medical Sciences, Kumamoto University, Japan
| | - Nicole Pui-Yu Ho
- Laboratory of Stem Cell Stress, International Research Center for Medical Sciences, Kumamoto University, Japan
| | - Hitoshi Takizawa
- Laboratory of Stem Cell Stress, International Research Center for Medical Sciences, Kumamoto University, Japan; Center for Metabolic Regulation of Healthy Aging, Kumamoto University, Japan.
| |
Collapse
|
22
|
Zhang H, Chen A, Li S, Chen K, You X, Bian Y, Li C, Liu S, Huang J, Zhang S. Spatio-Temporal Change of Skin and Oral Microbiota: A Longitudinal Study of Microbial Diversity and Stability. Electrophoresis 2025; 46:92-103. [PMID: 39800869 DOI: 10.1002/elps.202400160] [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: 07/26/2024] [Revised: 11/13/2024] [Accepted: 01/02/2025] [Indexed: 01/29/2025]
Abstract
The human skin and oral cavity harbor complex microbial communities, which exist in dynamic equilibrium with the host's physiological state and the external environment. This study investigates the microbial atlas of human skin and oral cavities using samples collected over a 10-month period, aiming to assess how both internal and external factors influence the human microbiome. We examined bacterial community diversity and stability across various body sites, including palm and nasal skin, saliva, and oral epithelial cells, during environmental changes and a COVID-19 pandemic. The skin microbiome was confirmed to display spatial and temporal stability compared to the oral microbiome, particularly the oral epithelium, which was susceptible to changes in the host's physiological state and immune response. Moreover, significant differences in the microbial community structure among the 4 sample types were observed, and 87 distinct bacteria biomarkers were identified. The random forest prediction model achieved an overall prediction accuracy of 95.24% across the four types of samples studied. Additionally, nasal skin samples showed significant promise for individual identification through profiling the skin microbiota. These findings highlight the potential of skin and oral microbiota as forensic markers for inferring body sites and identifying individuals. In summary, despite facing limitations such as a small cohort size and the need for broader validation, this research provides an overall perspective and initial insights for refining experimental designs and conducting in-depth research in various microbial research fields.
Collapse
Affiliation(s)
- Han Zhang
- Department of Forensic Medicine, Guizhou Medical University, Guiyang, Guizhou, P. R. China
- Institute of Forensic Science, Fudan University, Shanghai, P. R. China
| | - Anqi Chen
- Institute of Forensic Science, Fudan University, Shanghai, P. R. China
| | - Shilin Li
- Institute of Forensic Science, Fudan University, Shanghai, P. R. China
- Department of Anthropology and Human Genetics, School of Life Sciences, MOE Key Laboratory of Contemporary Anthropology, Fudan University, Shanghai, P. R. China
| | - Kaiqin Chen
- Key Laboratory of Cell Engineering of Guizhou Province, Clinical Stem Cell Research Institute, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou, P. R. China
| | - Xuechun You
- Institute of Forensic Science, Fudan University, Shanghai, P. R. China
| | - Yingnan Bian
- Enlight medical Technologies, Pudong New Area, Shanghai, P. R. China
| | - Chengtao Li
- Institute of Forensic Science, Fudan University, Shanghai, P. R. China
| | - Shiquan Liu
- Institute of Evidence Law and Forensic Science, China University of Political Science and Law, Beijing, P. R. China
| | - Jiang Huang
- The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, Guizhou, P. R. Chilna
| | - Suhua Zhang
- Institute of Forensic Science, Fudan University, Shanghai, P. R. China
| |
Collapse
|
23
|
Timmis K, Karahan ZC, Ramos JL, Koren O, Pérez‐Cobas AE, Steward K, de Lorenzo V, Caselli E, Douglas M, Schwab C, Rivero V, Giraldo R, Garmendia J, Turner RJ, Perlmutter J, Borrero de Acuña JM, Nikel PI, Bonnet J, Sessitsch A, Timmis JK, Pruzzo C, Prieto MA, Isazadeh S, Huang WE, Clarke G, Ercolini D, Häggblom M. Microbes Saving Lives and Reducing Suffering. Microb Biotechnol 2025; 18:e70068. [PMID: 39844583 PMCID: PMC11754571 DOI: 10.1111/1751-7915.70068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Accepted: 11/25/2024] [Indexed: 01/24/2025] Open
Affiliation(s)
- Kenneth Timmis
- Institute of MicrobiologyTechnical University BraunschweigBraunschweigGermany
| | - Zeynep Ceren Karahan
- Department of Medical Microbiology and Ibn‐i Sina Hospital Central Microbiology LaboratoryAnkara University School of MedicineAnkaraTurkey
| | - Juan Luis Ramos
- Consejo Superior de Investigaciones Científicas, Estación Experimental del ZaidínGranadaSpain
| | - Omry Koren
- Azrieli Faculty of MedicineBar‐Ilan UniversitySafedIsrael
| | - Ana Elena Pérez‐Cobas
- Department of Microbiology, Ramón y Cajal Institute for Health Research (IRYCIS)Ramón y Cajal University HospitalMadridSpain
- CIBER in Infectious Diseases (CIBERINFEC)MadridSpain
| | | | - Victor de Lorenzo
- Department of Systems BiologyNational Centre of Biotechnology CSICMadridSpain
| | - Elisabetta Caselli
- Section of Microbiology, Department of Environmental and Prevention SciencesUniversity of FerraraFerraraItaly
| | - Margaret Douglas
- Usher InstituteUniversity of Edinburgh Medical School, and Public Health ScotlandEdinburghUK
| | - Clarissa Schwab
- Department of Biological and Chemical EngineeringAarhus UniversityAarhusDenmark
| | - Virginia Rivero
- Polymer Biotechnology Lab, Biological Research Center Margarita SalasSpanish National Research Council (CIB‐CSIC)MadridSpain
| | - Rafael Giraldo
- Department of Microbial BiotechnologyNational Centre for Biotechnology (CNB‐CSIC)MadridSpain
| | - Junkal Garmendia
- Instituto de AgrobiotecnologíaConsejo Superior de Investigaciones Científicas (IdAB‐CSIC)‐Gobierno de Navarra, MutilvaMadridSpain
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES)MadridSpain
| | - Raymond J. Turner
- Department of Biological SciencesUniversity of CalgaryCalgaryAlbertaCanada
| | | | | | - Pablo Ivan Nikel
- The Novo Nordisk Foundation Center for BiosustainabilityTechnical University of DenmarkLyngbyDenmark
| | - Jerome Bonnet
- Centre de Biochimie Structurale, INSERM/CNRSUniversity of MontpellierMontpellierFrance
| | - Angela Sessitsch
- Bioresources UnitAIT Austrian Institute of TechnologyViennaAustria
| | - James K. Timmis
- Department of Political ScienceUniversity of FreiburgFreiburgGermany
- Athena Institute for Research on Innovation and Communication in Health and Life SciencesVrije UniversiteitAmsterdamThe Netherlands
| | - Carla Pruzzo
- Department of Earth, Environmental and Life Sciences (DISTAV)University of GenoaGenovaItaly
| | - M. Auxiliadora Prieto
- Polymer Biotechnology Lab, Biological Research Center Margarita SalasSpanish National Research Council (CIB‐CSIC)MadridSpain
| | - Siavash Isazadeh
- Corporate Technical & PerformanceVeolia North AmericaParamusNew JerseyUSA
| | - Wei E. Huang
- Department of Engineering ScienceUniversity of OxfordOxfordUK
| | - Gerard Clarke
- APC Microbiome IrelandUniversity College CorkCorkIreland
- Department of Psychiatry & Neurobehavioral SciencesUniversity College CorkCorkIreland
| | - Danilo Ercolini
- Department of Agricultural SciencesUniversity of Naples Federico IINaplesItaly
| | - Max Häggblom
- Department of Biochemistry and Microbiology, RutgersThe State University of New JerseyNew BrunswickNew JerseyUSA
| |
Collapse
|
24
|
Zhang T, Luo R, Ehrström M, Melican K. Staphylococcus lugdunensis does not exert competitive exclusion on human corneocytes. MICROBIOLOGY (READING, ENGLAND) 2025; 171. [PMID: 39888662 DOI: 10.1099/mic.0.001522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2025]
Abstract
Human skin is our primary physical barrier and largest immune organ, and it also hosts a protective microbiota. Staphylococci are prominent members of the skin microbiota, including the ubiquitous coagulase-negative staphylococci (CoNS). The coagulase-positive Staphylococcus aureus is found as part of the microbiota, but it poses clinical concern due to its potential pathogenicity and antibiotic resistance. Recently, a CoNS, Staphylococcus lugdunensis, has been shown to inhibit S. aureus growth via the production of a novel antibiotic, lugdunin. In this study, we use human skin models to understand the spatial relationships between the CoNS Staphylococcus epidermidis and S. lugdunensis with S. aureus during colonization of human skin. We investigated the attachment patterns of the bacteria, both individually and in competition. Surprisingly, we found that attachment did not always correlate with colonization ability. S. lugdunensis exhibited significantly reduced attachment to human skin stratum corneum but was an efficient longer-term colonizer. S. lugdunensis had a distinct attachment pattern on human corneocytes, with no significant overlap, or competitive exclusion, with the other strains. S. lugdunensis is a potential probiotic strain, with a proven ability to suppress S. aureus. Before this potential can be realized, however, further research is needed to understand how this strain adheres and interacts with other bacteria in the human skin microenvironment.
Collapse
Affiliation(s)
- Tianqi Zhang
- AIMES-Center for the Advancement of Integrated Medical and Engineering Sciences, Karolinska Institutet and KTH Royal Institute of Technology, SE-171 77 Stockholm, Sweden
- Department of Neuroscience, Karolinska Institutet, SE-171 77 Stockholm, Sweden
| | - Ran Luo
- AIMES-Center for the Advancement of Integrated Medical and Engineering Sciences, Karolinska Institutet and KTH Royal Institute of Technology, SE-171 77 Stockholm, Sweden
- Department of Neuroscience, Karolinska Institutet, SE-171 77 Stockholm, Sweden
| | | | - Keira Melican
- AIMES-Center for the Advancement of Integrated Medical and Engineering Sciences, Karolinska Institutet and KTH Royal Institute of Technology, SE-171 77 Stockholm, Sweden
- Department of Neuroscience, Karolinska Institutet, SE-171 77 Stockholm, Sweden
| |
Collapse
|
25
|
Chen YS, Chien AS, Li CC, Lin CC, Wu RJ. Effects of Commonly Used Vegetable Oils on Skin Barrier Function and Staphylococcus aureus Biofilm. J Oleo Sci 2025; 74:97-106. [PMID: 39756997 DOI: 10.5650/jos.ess24032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2025] Open
Abstract
Adding of vegetable oils to skincare products or the use of plant oils for oil care is a current trend. Therefore, the safety and functionality of vegetable oils are of great concern to consumers and cosmetics manufacturers. This study focused on three types of vegetable oils: sunflower oil (SO), andiroba oil (AO) and hydrogenated olive oil (HOO). We conducted a comprehensive evaluation of the oils, which encompassed their ability to protect mouse skin keratinocytes (XB-2) and mouse fibroblasts (NIH 3T3) from damage caused by the surfactant sodium lauryl sulfate (SLS), their influence on the levels of filaggrin and collagen, their potential to aid in wound healing, and their effectiveness in anti-Staphylococcus aureus biofilm formation. The results showed that SO, AO and HOO at a concentration of 1.5 × 10-4 % (v/v) have the ability to defend against SLS-induced cell damage, increase wound healing ability and the filaggrin and collagen content to XB-2 or NIH 3T3 cells. SO, AO and HOO at a concentration of 3.75 × 10-3 % also have the anti-biofilm ability. Among the oils, AO can inhibit S. aureus biofilm composed of either polysaccharides or proteins. Therefore, the tested vegetable oils and can be applied to the cosmetics field as ingredients to repair damaged skin and preserve skin barrier stability.
Collapse
Affiliation(s)
- Yi-Shyan Chen
- Department of Cosmetic Science, Providence University
| | - An-Sin Chien
- Department of Cosmetic Science, Providence University
| | - Chih-Ching Li
- Department of Applied Chemistry, Providence University
- Department of Pediatrics, Chang Bing Show Chwan Memorial Hospital
| | | | - Ren-Jang Wu
- Department of Applied Chemistry, Providence University
| |
Collapse
|
26
|
Li L, Du H, Meng C, Fan L, Liu H, Han X, Ge T, Su L, Yao X, Wang X. The relationship between indoor airborne culturable bacteria with passenger flow in 132 traffic stations during 2019-2020, China. JOURNAL OF ENVIRONMENTAL MANAGEMENT 2025; 373:123482. [PMID: 39612790 DOI: 10.1016/j.jenvman.2024.123482] [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: 05/08/2024] [Revised: 11/02/2024] [Accepted: 11/24/2024] [Indexed: 12/01/2024]
Abstract
This study aims to provide technical support for the development of a rational administration strategy for indoor environment and passenger flow of stations. The analysis was conducted monitor data of long distance bus stations and train/high speed railway stations from the National project. The monitor and surveyed data included indoor airborne culturable bacteria (IAB), passenger flow, area and height of stations. Data analysis involved the use of paired non-parametric tests, correlation analysis, and mixed linear regression methods. A total of 132 pairs of stations were examined in this study. Additionally, significant variations were observed in the monitoring results for the IAB and passenger flow between 2019 and 2020 (p < 0.05). Specifically, the median values of these two factors in 2020 decreased by 50.52% and 48.33%, respectively, compared to 2019. (p < 0.05). A positive relationship between the daily average passenger flow and the IAB in long distance bus stations and train stations, particularly pronounced with medium sized waiting rooms, which located in subtropical cities with a general level of economic development. The mixed effect regression analysis revealed a significant association between passenger flow (OR = 1.564, 1.288-1.898), per capita volume (OR = 0.856, 0.733-0.998), and per capita area (OR = 0.806, 0.678-0.956) with the IAB (p < 0.05) just in long distance bus stations. It is crucial to regulate passenger flow and per capita area in the waiting room of long distance bus stations to mitigate the spread of airborne culturable bacteria during infectious disease epidemics.
Collapse
Affiliation(s)
- Li Li
- China CDC Key Laboratory of Environment and Population Health, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Hang Du
- China CDC Key Laboratory of Environment and Population Health, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Chong Meng
- China architecture design and research group, Beijing, China
| | - Lin Fan
- China CDC Key Laboratory of Environment and Population Health, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Hang Liu
- China CDC Key Laboratory of Environment and Population Health, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Xu Han
- China CDC Key Laboratory of Environment and Population Health, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Tanxi Ge
- China CDC Key Laboratory of Environment and Population Health, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Liqin Su
- China CDC Key Laboratory of Environment and Population Health, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Xiaoyuan Yao
- China CDC Key Laboratory of Environment and Population Health, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Xianliang Wang
- China CDC Key Laboratory of Environment and Population Health, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing, China.
| |
Collapse
|
27
|
Smith A, Dumbrava R, Ghori NUH, Foster R, Campbell J, Duthie A, Hoyne G, Rademaker M, Bowen AC. An Overview of the Skin Microbiome, the Potential for Pathogen Shift, and Dysbiosis in Common Skin Pathologies. Microorganisms 2025; 13:54. [PMID: 39858822 PMCID: PMC11767775 DOI: 10.3390/microorganisms13010054] [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: 12/05/2024] [Revised: 12/22/2024] [Accepted: 12/27/2024] [Indexed: 01/27/2025] Open
Abstract
Recent interest in the diverse ecosystem of bacteria, fungi, parasites, and viruses that make up the skin microbiome has led to several studies investigating the microbiome in healthy skin and in a variety of dermatological conditions. An imbalance of the normal skin flora can cause some skin diseases, and current culture techniques are often unable to detect a microorganism to further our understanding of the clinical-microbiological correlates of disease and dysbiosis. Atopic dermatitis and rosacea are presentations that GPs often manage that may have an infective or microbiological component and can be challenging to treat. We aim to discuss the implications of the skin microbiome including the impact of dysbiosis on conditions such as these. We will also discuss some clinical pearls for initial and future directions of the management of conditions such as atopic dermatitis, rosacea, and hidradenitis suppurativa. Further research using culture-independent techniques is needed for conditions involving microbial dysbiosis to advance our knowledge of skin disease pathophysiology and guide future management.
Collapse
Affiliation(s)
- Anita Smith
- Healthy Skin Team, Wesfarmers Centre of Vaccines and Infectious Diseases, The Kids Research Institute Australia, Perth, WA 6009, Australia
- Perth Children’s Hospital, Perth, WA 6009, Australia
- School of Medicine, University of Notre Dame, Fremantle, WA 6160, Australia
- Sir Charles Gairdner Hospital, Perth, WA 6009, Australia
- Department of Dermatology, Fiona Stanley Hospital, Perth, WA 6150, Australia
| | | | - Noor-Ul-Huda Ghori
- Healthy Skin Team, Wesfarmers Centre of Vaccines and Infectious Diseases, The Kids Research Institute Australia, Perth, WA 6009, Australia
- Division of Infection and Immunity, School of Biomedical Sciences, University of Western Australia, Perth, WA 6009, Australia
| | - Rachael Foster
- Healthy Skin Team, Wesfarmers Centre of Vaccines and Infectious Diseases, The Kids Research Institute Australia, Perth, WA 6009, Australia
- Perth Children’s Hospital, Perth, WA 6009, Australia
- Sir Charles Gairdner Hospital, Perth, WA 6009, Australia
- Department of Dermatology, Fiona Stanley Hospital, Perth, WA 6150, Australia
| | | | - Andrew Duthie
- Central Perth Skin Clinic, Perth, WA 6000, Australia
| | - Gerard Hoyne
- Institute of Respiratory Health, QEII Medical Centre, Perth, WA 6009, Australia
- School of Health Sciences, University of Notre Dame, Fremantle, WA 6160, Australia
| | | | - Asha C. Bowen
- Healthy Skin Team, Wesfarmers Centre of Vaccines and Infectious Diseases, The Kids Research Institute Australia, Perth, WA 6009, Australia
- Perth Children’s Hospital, Perth, WA 6009, Australia
- School of Medicine, University of Notre Dame, Fremantle, WA 6160, Australia
- Division of Paediatrics, School of Medicine, University of Western Australia, Perth, WA 6009, Australia
| |
Collapse
|
28
|
Lin YH, Chen YC, Yii CY, Yong SB. Temporal Relationships Between Staphylococcus aureus Colonization, Filaggrin Expression, and Pediatric Atopic Dermatitis-Correspondence. Int J Rheum Dis 2025; 28:e70071. [PMID: 39791552 DOI: 10.1111/1756-185x.70071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Accepted: 01/05/2025] [Indexed: 01/12/2025]
Affiliation(s)
- Yu-Hsi Lin
- School of Chinese Medicine, China Medical University, Taichung, Taiwan
| | - Yi-Chen Chen
- School of Medicine, China Medical University, Taichung, Taiwan
| | - Chin-Yuan Yii
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Landseed International Hospital, Taoyuan, Taiwan
- Department of Biomedical Sciences and Engineering, National Central University, Taoyuan, Taiwan
| | - Su-Boon Yong
- Center for Allergy, Immunology, and Microbiome (AIM), China Medical University Hospital, Taichung, Taiwan
- Department of Allergy, Immunology, and Rheumatology (AIR), China Medical University Children's Hospital, Taichung, Taiwan
- Department of Medicine, College of Medicine, China Medical University, Taichung, Taiwan
| |
Collapse
|
29
|
Manus MB, Sardaro MLS, Dada O, Davis MI, Romoff MR, Torello SG, Ubadigbo E, Wu RC, Miller ES, Amato KR. Interactions with alloparents are associated with the diversity of infant skin and fecal bacterial communities in Chicago, United States. Am J Hum Biol 2025; 37:e23972. [PMID: 37632331 PMCID: PMC11667966 DOI: 10.1002/ajhb.23972] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 07/20/2023] [Accepted: 07/21/2023] [Indexed: 08/28/2023] Open
Abstract
INTRODUCTION Social interactions shape the infant microbiome by providing opportunities for caregivers to spread bacteria through physical contact. With most research focused on the impact of maternal-infant contact on the infant gut microbiome, it is unclear how alloparents (i.e., caregivers other than the parents) influence the bacterial communities of infant body sites that are frequently contacted during bouts of caregiving, including the skin. METHODS To begin to understand how allocare may influence the diversity of the infant microbiome, detailed questionnaire data on infant-alloparent relationships and specific allocare behaviors were coupled with skin and fecal microbiome samples (four body sites) from 48 infants living in Chicago, United States. RESULTS Data from 16S rRNA gene amplicon sequencing indicated that infant skin and fecal bacterial diversity showed strong associations (positive and negative) to having female adult alloparents. Alloparental feeding and co-sleeping displayed stronger associations to infant bacterial diversity compared to playing or holding. The associations with allocare behaviors differed in magnitude and direction across infant body sites. Bacterial relative abundances varied by infant-alloparent relationship and breastfeeding status. CONCLUSION This study provides some of the first evidence of an association between allocare and infant skin and fecal bacterial diversity. The results suggest that infants' exposure to bacteria from the social environment may vary based on infant-alloparent relationships and allocare behaviors. Since the microbiome influences immune system development, variation in allocare that impacts the diversity of infant bacterial communities may be an underexplored dimension of the social determinants of health in early life.
Collapse
Affiliation(s)
- Melissa B. Manus
- Department of AnthropologyNorthwestern UniversityEvanstonIllinoisUSA
| | - Maria Luisa Savo Sardaro
- Department of AnthropologyNorthwestern UniversityEvanstonIllinoisUSA
- Department of Human Science and Promotion of the Quality of LifeUniversity of San RaffaeleRomeItaly
| | - Omolola Dada
- Department of AnthropologyNorthwestern UniversityEvanstonIllinoisUSA
| | - Maya I. Davis
- Department of AnthropologyNorthwestern UniversityEvanstonIllinoisUSA
| | - Melissa R. Romoff
- Department of AnthropologyNorthwestern UniversityEvanstonIllinoisUSA
| | | | - Esther Ubadigbo
- Department of AnthropologyNorthwestern UniversityEvanstonIllinoisUSA
| | - Rebecca C. Wu
- Department of AnthropologyNorthwestern UniversityEvanstonIllinoisUSA
| | - Emily S. Miller
- Department of Obstetrics and Gynecology, Division of Maternal Fetal MedicineFeinberg School of Medicine, Northwestern UniversityChicagoIllinoisUSA
| | | |
Collapse
|
30
|
Caukwell J, Assenza S, Hassan KA, Neilan BA, Clulow AJ, Salvati Manni L, Fong WK. Lipidic drug delivery systems are responsive to the human microbiome. J Colloid Interface Sci 2025; 677:293-302. [PMID: 39146817 DOI: 10.1016/j.jcis.2024.07.216] [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: 07/15/2024] [Accepted: 07/27/2024] [Indexed: 08/17/2024]
Abstract
In vitro and in vivo tests for therapeutic agents are typically conducted in sterile environments, but many target areas for drug delivery are home to thousands of microbial species. Here, we examine the behaviour of lipidic nanomaterials after exposure to representative strains of four bacterial species found in the gastrointestinal tract and skin. Small angle X-ray scattering measurements show that the nanostructure of monoolein cubic and inverse hexagonal phases are transformed, respectively, into inverse hexagonal and inverse micellar cubic phases upon exposure to a strain of live Staphylococcus aureus often present on skin and mucosa. Further investigation demonstrates that enzymatic hydrolysis and cell membrane lipid transfer are both likely responsible for this effect. The structural responses to S. aureus are rapid and significantly reduce the rate of drug release from monoolein-based nanomaterials. These findings are the first to demonstrate how a key species in the live human microbiome can trigger changes in the structure and drug release properties of lipidic nanomaterials. The effect appears to be strain specific, varies from patient to patient and body region to body region, and is anticipated to affect the bioapplication of monoglyceride-based formulations.
Collapse
Affiliation(s)
- Jonathan Caukwell
- School of Chemistry, Monash University, Clayton 3800, VIC, Australia; School of Environmental and Life Sciences, University of Newcastle, Callaghan 2308, NSW, Australia
| | - Salvatore Assenza
- Departamento de Física Teórica de la Materia Condensada, Universidad Autónoma de Madrid, E-28049 Madrid, Spain; Condensed Matter Physics Center (IFIMAC), Universidad Autónoma de Madrid, E-28049 Madrid, Spain; Instituto Nicolás Cabrera, Universidad Autónoma de Madrid, E-28049 Madrid, Spain
| | - Karl A Hassan
- School of Environmental and Life Sciences, University of Newcastle, Callaghan 2308, NSW, Australia
| | - Brett A Neilan
- School of Environmental and Life Sciences, University of Newcastle, Callaghan 2308, NSW, Australia
| | - Andrew J Clulow
- Australian Synchrotron, ANSTO, 800 Blackburn Road, Clayton, VIC 3168, Australia; Drug Delivery, Disposition & Dynamics, Monash Institute of Pharmaceutical Sciences, 381 Royal Parade, Parkville, VIC 3052, Australia
| | - Livia Salvati Manni
- School of Chemistry, Monash University, Clayton 3800, VIC, Australia; Australian Synchrotron, ANSTO, 800 Blackburn Road, Clayton, VIC 3168, Australia; School of Chemistry and University of Sydney Nano Institute, The University of Sydney, NSW 2006, Australia.
| | - Wye-Khay Fong
- School of Chemistry, Monash University, Clayton 3800, VIC, Australia; School of Environmental and Life Sciences, University of Newcastle, Callaghan 2308, NSW, Australia.
| |
Collapse
|
31
|
Zhang J, Feng Y, Li D, Shi D. Fungal influence on immune cells and inflammatory responses in the tumor microenvironment (Review). Oncol Lett 2025; 29:50. [PMID: 39564373 PMCID: PMC11574707 DOI: 10.3892/ol.2024.14796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Accepted: 07/25/2024] [Indexed: 11/21/2024] Open
Abstract
In recent years, a growing body of research has highlighted the significant influence of the microbiota on tumor immunity within the tumor microenvironment (TME). While much attention has been given to bacteria, emerging evidence suggests that fungi also play crucial roles in tumor development. The present review aimed to consolidate the latest findings on the mechanisms governing the interactions between fungi and the immune system or TME. By elucidating these intricate mechanisms, novel insights into the modulation of tumor immunity and therapeutic strategies may be uncovered. Ultimately, a deeper understanding of the interplay between fungi and the TME holds promise for the development of innovative management strategies and targeted drugs to enhance tumor therapy efficacy.
Collapse
Affiliation(s)
- Jinke Zhang
- Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, P.R. China
- Department of Microbiology and Immunology, Georgetown University Medical Center, Washington DC 20057, USA
| | - Yahui Feng
- Laboratory of Medical Mycology, Jining No. 1 People's Hospital, Jining, Shandong 272001, P.R. China
| | - Dongmei Li
- Department of Microbiology and Immunology, Georgetown University Medical Center, Washington DC 20057, USA
| | - Dongmei Shi
- Laboratory of Medical Mycology, Jining No. 1 People's Hospital, Jining, Shandong 272001, P.R. China
- Department of Dermatology, Jining No. 1 People's Hospital, Jining, Shandong 272001, P.R. China
| |
Collapse
|
32
|
Pakkiyam S, Marimuthu M, Kumar J, Ganesh V, Veerapandian M. Microbial crosstalk with dermal immune system: A review on emerging analytical methods for macromolecular detection and therapeutics. Int J Biol Macromol 2024; 293:139369. [PMID: 39743089 DOI: 10.1016/j.ijbiomac.2024.139369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 12/24/2024] [Accepted: 12/29/2024] [Indexed: 01/04/2025]
Abstract
According to global health metrics, clinical symptoms such as cellulitis and pyoderma associated with skin diseases are a significant burden worldwide, affecting 2.2 million disability-adjusted life years in 2020. There is a strong correlation between the commensal bacteria and the host immune system. Classical methods deployed in dermal biofilm crosstalk studies often hamper many individuals from early diagnosis and rationalized therapy. Herein, the present report aims to study the role of skin microbiota and mechanisms of microbial crosstalk with host immune system. The emerging analytical tools devised for sensor/biosensor platforms, including molecularly imprinted polymers, microarrays, aptamers, CRISPR-cas9, and optical/electrochemical approaches, are discussed as alternative methods for important biomarker analysis. Further, the types and characteristics of microorganism-derived macromolecules and the recent skin organoid toward personalized therapy are highlighted. This information will largely benefit researchers involved in the pathophysiology of skin disease, wound dressing materials, including diagnostic and healing patch designs, in addition to biological macromolecules devoted to wound repair.
Collapse
Affiliation(s)
- Sangavi Pakkiyam
- Electrodics and Electrocatalysis Division, CSIR-Central Electrochemical Research Institute (CECRI), Karaikudi 630 003, India; Academy of Scientific & Innovative Research (AcSIR), Ghaziabad 201 002, India
| | - Mohana Marimuthu
- Department of Biotechnology, Faculty of Engineering and Technology, SRM Institute of Science and Technology Tiruchirappalli Campus, Trichy 621 105, Tamil Nadu, India; Innovaspark STEM Edutainment Centre, Karaikudi 630 003, Tamil Nadu, India
| | - Jitendra Kumar
- Nuclear Agriculture and Biotechnology Division, Bhabha Atomic Research Centre, Mumbai 400 085, India; Homi Bhabha National Institute (HBNI), Anushaktinagar, Mumbai 400 094, India
| | - V Ganesh
- Electrodics and Electrocatalysis Division, CSIR-Central Electrochemical Research Institute (CECRI), Karaikudi 630 003, India; Academy of Scientific & Innovative Research (AcSIR), Ghaziabad 201 002, India.
| | - Murugan Veerapandian
- Electrodics and Electrocatalysis Division, CSIR-Central Electrochemical Research Institute (CECRI), Karaikudi 630 003, India; Academy of Scientific & Innovative Research (AcSIR), Ghaziabad 201 002, India.
| |
Collapse
|
33
|
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
|
34
|
Assaf S, Kelly O. Nutritional Dermatology: Optimizing Dietary Choices for Skin Health. Nutrients 2024; 17:60. [PMID: 39796494 PMCID: PMC11723311 DOI: 10.3390/nu17010060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2024] [Revised: 12/19/2024] [Accepted: 12/25/2024] [Indexed: 01/13/2025] Open
Abstract
BACKGROUND/OBJECTIVES Youthful, smooth skin is highly desired in modern society. Individuals invest in cosmetics, plastic surgeons, and dermatologists in pursuit of perfect skin. However, many do not seek out dietary changes to improve skin health. Although research has been conducted on the role of nutrition and select nutrients and phytonutrients on skin health, there is a lack of healthy food recommendations for clear skin. METHODS The literature was assessed to determine which nutrients and phytonutrients play a significant role in the protection and maintenance of skin health. Key compounds were highlighted as there is evidence to suggest they have a significant role in skin health: vitamin A, vitamin C, vitamin D, vitamin E, zinc, omega-6 and omega-3 fatty acids, polyphenols/flavonoids, copper, selenium, and silicon. USDA FoodData Central and FooDB (food database), were utilized to select foods and food groups containing the key nutrients and phytonutrients. RESULTS A skin-healthy dietary pattern is proposed in addition to a scoring system to assess diet. A sample skin-healthy daily diet was designed, using only whole foods, that met the Daily Values for vitamins and minerals and contained key compounds for skin health. CONCLUSIONS There is a clear link between nutrition and skin health, or nutritional dermatology; however, more research needs to be done to find the intersection between both disciplines.
Collapse
Affiliation(s)
- Sandi Assaf
- Department of Molecular and Cellular Biology, College of Osteopathic Medicine, Sam Houston State University, Conroe, TX 77304, USA;
| | | |
Collapse
|
35
|
Wagner N, Valeriano VD, Diou-Hirtz S, Björninen E, Åkerström U, Engstrand L, Schuppe-Koistinen I, Gillbro JM. Microbial Dynamics: Assessing Skincare Regimens' Impact on the Facial Skin Microbiome and Skin Health Parameters. Microorganisms 2024; 12:2655. [PMID: 39770857 PMCID: PMC11728252 DOI: 10.3390/microorganisms12122655] [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: 11/25/2024] [Revised: 12/06/2024] [Accepted: 12/19/2024] [Indexed: 01/16/2025] Open
Abstract
The human skin microbiome, a complex ecosystem of microbes, plays a pivotal role in skin health. This study aimed to investigate the impact of two skincare regimens, with preservatives (CSPs) and preservative-free (PFPs), on the skin microbiome in correlation to skin quality. double-blind randomized cosmetic studywith a split-face design was conducted on 26 female participants. Microbial diversity and abundance were analyzed using 16S rRNA amplicon sequence data and skin quality utilizing the Antera 3D skin camera. We confirmed earlier studies on the identification of major skin microbial taxa at the genus level, including Cutibacterium acnes, Corynebacterium, and Neisseriaceae as a predominant part of the facial skin microbiome. Furthermore, microbiome profile-based subgrouping was employed, which revealed that the cluster, characterized by the Neisseriaceae family as its predominant organism, exhibited significant reduction in folds count, fine lines, and redness after application of PFP compared to CSP. A Spearman correlation analysis highlighted the correlation between changes in specific bacteria and skin quality parameters such as redness, pores, and texture in the context of comparing PFP and CSP. Overall, the PFP treatment demonstrated a greater number of significant correlations between bacterial changes and skin quality compared to the CSP treatment, suggesting a distinct impact of the preservative-free skincare regimen on the skin microbiome and skin quality. Our study provides insights into different microbiome-centered approaches to improve our understanding of the skin microbiome's interplay with skin quality but also highlights the need for larger, comprehensive research to further understand the microbiome's role in dermatology.
Collapse
Affiliation(s)
- Nicole Wagner
- Center for Translational Microbiome Research, CTMR, Department of Microbiology, Tumor and Cell Biology (MTC), Science for Life Laboratory, Karolinska Institute, 171 65 Solna, Sweden; (V.D.V.); (S.D.-H.); (E.B.); (L.E.); (I.S.-K.)
| | - Valerie Diane Valeriano
- Center for Translational Microbiome Research, CTMR, Department of Microbiology, Tumor and Cell Biology (MTC), Science for Life Laboratory, Karolinska Institute, 171 65 Solna, Sweden; (V.D.V.); (S.D.-H.); (E.B.); (L.E.); (I.S.-K.)
| | - Samuel Diou-Hirtz
- Center for Translational Microbiome Research, CTMR, Department of Microbiology, Tumor and Cell Biology (MTC), Science for Life Laboratory, Karolinska Institute, 171 65 Solna, Sweden; (V.D.V.); (S.D.-H.); (E.B.); (L.E.); (I.S.-K.)
| | - Evelina Björninen
- Center for Translational Microbiome Research, CTMR, Department of Microbiology, Tumor and Cell Biology (MTC), Science for Life Laboratory, Karolinska Institute, 171 65 Solna, Sweden; (V.D.V.); (S.D.-H.); (E.B.); (L.E.); (I.S.-K.)
| | - Ulf Åkerström
- Skinome Research AB, Hornsgatan 172, 117 28 Stockholm, Sweden;
| | - Lars Engstrand
- Center for Translational Microbiome Research, CTMR, Department of Microbiology, Tumor and Cell Biology (MTC), Science for Life Laboratory, Karolinska Institute, 171 65 Solna, Sweden; (V.D.V.); (S.D.-H.); (E.B.); (L.E.); (I.S.-K.)
| | - Ina Schuppe-Koistinen
- Center for Translational Microbiome Research, CTMR, Department of Microbiology, Tumor and Cell Biology (MTC), Science for Life Laboratory, Karolinska Institute, 171 65 Solna, Sweden; (V.D.V.); (S.D.-H.); (E.B.); (L.E.); (I.S.-K.)
| | | |
Collapse
|
36
|
Zhu W, Wang X, He D, Chen B. Causal relationships between skin microbiome and pathological scars: a bidirectional mendelian randomization study. Arch Dermatol Res 2024; 317:139. [PMID: 39704837 DOI: 10.1007/s00403-024-03576-y] [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: 10/02/2024] [Revised: 10/25/2024] [Accepted: 11/26/2024] [Indexed: 12/21/2024]
Abstract
Skin bacteria infection could be a potential risk factor on wound scar formation, yet the specifics of this relationship are not fully understood. This research investigates the causal relationships between specific skin microbiome and these diseases by using bidirectional Mendelian randomization (MR). This study employed a bidirectional MR analysis using genome-wide association study (GWAS) data to analyze the associations between skin microbiome and pathological scar. Single nucleotide polymorphisms (SNPs) served as instrumental variables (IVs) in MR methods, including inverse variance weighted (IVW), and MR Egger. The IVW analysis suggested a significant relationship between specific skin microbiome and pathlogical scars. Actinomycetales_Sebaceous, Proteobacteria_Sebaceous, ASV072[Paracoccus (unc.)]_Dry, ASV008[Diaphorobacter nitroreducens]_Dry, Pseudomonadales_moist, ASV001[Propionibacterium acnes]_Moist, Moraxellaceae_moist, Flavobacteriaceae_Dry were significantly associated with keloid. Chryseobacterium_Moist, ASV016[Enhydrobacter(unc.)]_Moist, ASV021[Micrococcus(unc.)]_Moist, ASV022[Streptococcus salivarius]_Moist, Rhodobacteraceae_Dry, Staphylococcus_Moist, Micrococcaceae_Moist, ASV007[Anaerococcus(unc.)]_Dry, Betaproteobacteria_Moist and ASV001[Propionibacterium acnes]_Moist were significantly associated with hypertrophic scarring. Reverse MR analysis indicates both keloid and hypertrophic scar regulated the composition of the skin microbiome. The study revealed a possible correlation between some specific skin microbiome and pathlogical scars. Understanding these inverse relationship could help improve clinical treatment and reducing pathological scar formation.
Collapse
Affiliation(s)
- Weiwen Zhu
- Plastic surgery department, Fujian children's hospital, Fuzhou, China
| | - Xueming Wang
- Plastic surgery department, Fujian children's hospital, Fuzhou, China
| | - Derong He
- Plastic surgery department, Fujian children's hospital, Fuzhou, China
| | - Baiye Chen
- Plastic surgery department, Fujian children's hospital, Fuzhou, China.
| |
Collapse
|
37
|
Lesiak A, Paprocka P, Wnorowska U, Mańkowska A, Król G, Głuszek K, Piktel E, Spałek J, Okła S, Fiedoruk K, Durnaś B, Bucki R. Significance of host antimicrobial peptides in the pathogenesis and treatment of acne vulgaris. Front Immunol 2024; 15:1502242. [PMID: 39744637 PMCID: PMC11688235 DOI: 10.3389/fimmu.2024.1502242] [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: 09/26/2024] [Accepted: 12/02/2024] [Indexed: 01/11/2025] Open
Abstract
Acne vulgaris (AV) is a chronic inflammatory condition of the pilosebaceous units characterized by multiple immunologic, metabolic, hormonal, genetic, psycho-emotional dysfunctions, and skin microbiota dysbiosis. The latter is manifested by a decreased population (phylotypes, i.e., genetically distinct bacterial subgroups that play different roles in skin health and disease) diversity of the predominant skin bacterial commensal - Cutinbacterium acnes. Like in other dysbiotic disorders, an elevated expression of endogenous antimicrobial peptides (AMPs) is a hallmark of AV. AMPs, such as human β-defensins, cathelicidin LL-37, dermcidin, or RNase-7, due to their antibacterial and immunomodulatory properties, function as the first line of defense and coordinate the host-microbiota interactions. Therefore, AMPs are potential candidates for pharmaceutical prophylaxis or treating this condition. This study outlines the current knowledge regarding the importance of AMPs in AV pathomechanism in light of recent transcriptomic studies. In particular, their role in improving the tight junctions (TJs) skin barrier by activating the fundamental cellular proteins, such as PI3K, GSK-3, aPKC, and Rac1, is discussed. We hypothesized that the increased expression of AMPs and their patterns in AV act as a compensatory mechanism to protect the skin with an impaired permeability barrier. Therefore, AMPs could be key determinants in regulating AV development and progression, linking acne-associated immune responses and metabolic factors, like insulin/IGF-1 and PI3K/Akt/mTOR/FoxO1 signaling pathways or glucotoxicity. Research and development of anti-acne AMPs are also addressed.
Collapse
Affiliation(s)
- Agata Lesiak
- Institute of Medical Science, Collegium Medicum, Jan Kochanowski University in Kielce, Kielce, Poland
| | - Paulina Paprocka
- Institute of Medical Science, Collegium Medicum, Jan Kochanowski University in Kielce, Kielce, Poland
| | - Urszula Wnorowska
- Department of Medical Microbiology and Nanobiomedical Engineering, Medical University of Białystok, Białystok, Poland
| | - Angelika Mańkowska
- Institute of Medical Science, Collegium Medicum, Jan Kochanowski University in Kielce, Kielce, Poland
| | - Grzegorz Król
- Institute of Medical Science, Collegium Medicum, Jan Kochanowski University in Kielce, Kielce, Poland
| | - Katarzyna Głuszek
- Institute of Medical Science, Collegium Medicum, Jan Kochanowski University in Kielce, Kielce, Poland
| | - Ewelina Piktel
- Independent Laboratory of Nanomedicine, Medical University of Białystok, Białystok, Poland
| | - Jakub Spałek
- Institute of Medical Science, Collegium Medicum, Jan Kochanowski University in Kielce, Kielce, Poland
- Department of Otolaryngology, Holy-Cross Oncology Center of Kielce, Head and Neck Surgery, Kielce, Poland
| | - Sławomir Okła
- Institute of Medical Science, Collegium Medicum, Jan Kochanowski University in Kielce, Kielce, Poland
- Department of Otolaryngology, Holy-Cross Oncology Center of Kielce, Head and Neck Surgery, Kielce, Poland
| | - Krzysztof Fiedoruk
- Department of Medical Microbiology and Nanobiomedical Engineering, Medical University of Białystok, Białystok, Poland
| | - Bonita Durnaś
- Institute of Medical Science, Collegium Medicum, Jan Kochanowski University in Kielce, Kielce, Poland
- Department of Clinical Microbiology, Holy-Cross Oncology Center of Kielce, Kielce, Poland
| | - Robert Bucki
- Department of Medical Microbiology and Nanobiomedical Engineering, Medical University of Białystok, Białystok, Poland
| |
Collapse
|
38
|
Todorov SD, Tagg J, Algburi A, Tiwari SK, Popov I, Weeks R, Mitrokhin OV, Kudryashov IA, Kraskevich DA, Chikindas ML. The Hygienic Significance of Microbiota and Probiotics for Human Wellbeing. Probiotics Antimicrob Proteins 2024:10.1007/s12602-024-10419-9. [PMID: 39688648 DOI: 10.1007/s12602-024-10419-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/01/2024] [Indexed: 12/18/2024]
Abstract
The human body can be viewed as a combination of ecological niches inhabited by trillions of bacteria, viruses, fungi, and parasites, all united by the microbiota concept. Human health largely depends on the nature of these relationships and how they are built and maintained. However, personal hygiene practices have historically been focused on the wholesale elimination of pathogens and "hygiene-challenging microorganisms" without considering the collateral damage to beneficial and commensal species. The microbiota can vary significantly in terms of the qualitative and quantitative composition both between different people and within one person during life, and the influence of various environmental factors, including age, nutrition, bad habits, genetic factors, physical activity, medication, and hygienic practices, facilitates these changes. Disturbance of the microbiota is a predisposing factor for the development of diseases and also greatly influences the course and severity of potential complications. Therefore, studying the composition of the microbiota of the different body systems and its appropriate correction is an urgent problem in the modern world. The application of personal hygiene products or probiotics must not compromise health through disruption of the healthy microbiota. Where changes in the composition or metabolic functions of the microbiome may occur, they must be carefully evaluated to ensure that essential biological functions are unaffected. As such, the purpose of this review is to consider the microbiota of each of the "ecological niches" of the human body and highlight the importance of the microbiota in maintaining a healthy body as well as the possibility of its modulation through the use of probiotics for the prevention and treatment of certain human diseases.
Collapse
Affiliation(s)
- Svetoslav D Todorov
- ProBacLab, Laboratório de Microbiologia de Alimentos, Departamento de Alimentos E Nutrição Experimental, Food Research Center, Faculdade de Ciências Farmacêuticas, Universidade de São Paulo, São Paulo, SP, 05508-000, Brazil.
- Instituto Politécnico de Viana Do Castelo, 4900-347, Viana Do Castelo, Portugal.
| | - John Tagg
- Blis Technologies, South Dunedin, 9012, New Zealand
| | - Ammar Algburi
- Department of Microbiology, College of Veterinary Medicine, University of Diyala, Baqubah, Iraq
| | - Santosh Kumar Tiwari
- Department of Genetics, Maharshi Dayanand University, Rohtak, 124001, Haryana, India
| | - Igor Popov
- Center for Agrobiotechnology, Don State Technical University, Gagarina Sq., 1344002, Rostov-On-Don, Russia
- Division of Immunobiology and Biomedicine, Center of Genetics and Life Sciences, Sirius University of Science and Technology, Federal Territory Sirius, Olimpijskij Ave., 1, Federal Territory Sirius, Sirius, 354340, Russia
- Centre for Healthy Eating & Food Innovation (HEFI), Maastricht University-Campus Venlo, Villafloraweg, 1, 5928 SZ, Venlo, The Netherlands
| | - Richard Weeks
- Health Promoting Naturals Laboratory, School of Environmental and Biological Sciences, Rutgers, the State University of New Jersey, New Brunswick, NJ, 08901, USA
| | - Oleg V Mitrokhin
- Department of General Hygiene, I.M. Sechenov First Moscow State Medical University, Moscow, 119435, Russia
| | - Ilya A Kudryashov
- Department of General Hygiene, I.M. Sechenov First Moscow State Medical University, Moscow, 119435, Russia
| | - Denis A Kraskevich
- Department of General Hygiene, I.M. Sechenov First Moscow State Medical University, Moscow, 119435, Russia
| | - Michael L Chikindas
- Center for Agrobiotechnology, Don State Technical University, Gagarina Sq., 1344002, Rostov-On-Don, Russia.
- Health Promoting Naturals Laboratory, School of Environmental and Biological Sciences, Rutgers, the State University of New Jersey, New Brunswick, NJ, 08901, USA.
- Department of General Hygiene, I.M. Sechenov First Moscow State Medical University, Moscow, 119435, Russia.
| |
Collapse
|
39
|
Kaplan JB, Assa M, Mruwat N, Sailer M, Regmi S, Kridin K. Facultatively Anaerobic Staphylococci Enable Anaerobic Cutibacterium Species to Grow and Form Biofilms Under Aerobic Conditions. Microorganisms 2024; 12:2601. [PMID: 39770803 PMCID: PMC11678309 DOI: 10.3390/microorganisms12122601] [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: 11/20/2024] [Revised: 12/09/2024] [Accepted: 12/13/2024] [Indexed: 01/11/2025] Open
Abstract
Facultatively anaerobic Staphylococcus spp. and anaerobic Cutibacterium spp. are among the most prominent bacteria on human skin. Although skin microbes generally grow as multispecies biofilms, few studies have investigated the interaction between staphylococci and Cutibacterium spp. in dual-species biofilms. Here, we measured the mono- and dual-species biofilm formation of four staphylococcal species (S. epidermidis, S. hominis, S. capitis, and S. aureus) and two Cutibacterium spp. (C. acnes and C. avidum) cultured in vitro under both aerobic and anaerobic conditions. The biofilms were quantitated by rinsing them to remove planktonic cells, detaching the biofilm bacteria via sonication, and enumerating the cells by dilution plating. When cultured alone, staphylococci formed biofilms under both aerobic and anaerobic conditions, whereas Cutibacterium spp. formed biofilms only under anaerobic conditions. In co-culture, staphylococcal biofilm formation was unaffected by the presence of Cutibacterium spp., regardless of oxygen availability. However, Cutibacterium spp. biofilm formation was significantly enhanced in the presence of staphylococci, enabling robust growth under both anaerobic and aerobic conditions. Fluorescence confocal microscopy of the aerobic dual-species biofilms suggested that staphylococci create anaerobic niches at the base of the biofilm where C. acnes can grow. These findings demonstrate that staphylococci facilitate the colonization of Cutibacterium spp. in oxygen-rich environments, potentially explaining their presence in high numbers on the oxygen-exposed stratum corneum.
Collapse
Affiliation(s)
- Jeffrey B. Kaplan
- Laboratory for Skin Research, Institute for Medical Research, Galilee Medical Center, Nahariya 2210001, Israel; (N.M.); (K.K.)
| | - Michael Assa
- The Azrieli Faculty of Medicine, Bar-Ilan University, Safed 1311502, Israel;
| | - Noor Mruwat
- Laboratory for Skin Research, Institute for Medical Research, Galilee Medical Center, Nahariya 2210001, Israel; (N.M.); (K.K.)
| | - Miloslav Sailer
- Kane Biotech Inc., Winnipeg, MB R3T 6G2, Canada; (M.S.); (S.R.)
| | - Suresh Regmi
- Kane Biotech Inc., Winnipeg, MB R3T 6G2, Canada; (M.S.); (S.R.)
| | - Khalaf Kridin
- Laboratory for Skin Research, Institute for Medical Research, Galilee Medical Center, Nahariya 2210001, Israel; (N.M.); (K.K.)
- The Azrieli Faculty of Medicine, Bar-Ilan University, Safed 1311502, Israel;
| |
Collapse
|
40
|
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
|
41
|
Chen B, Yao L, Cai R, Chen W, Wang Y. Antimicrobial Resistance Profiles and mupA Gene Characterization of Staphylococcus epidermidis Recovered from Facial Skin of Healthy Females in Shanghai, China. Clin Cosmet Investig Dermatol 2024; 17:2813-2821. [PMID: 39677851 PMCID: PMC11639884 DOI: 10.2147/ccid.s481517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Accepted: 11/25/2024] [Indexed: 12/17/2024]
Abstract
Purpose To explore antimicrobial resistance profiles and mupA gene characterization of Staphylococcus epidermidis recovered from facial skin of healthy females in Shanghai, China. Patients and Methods In this study, we collected facial skin samples from 107 healthy females in Shanghai, China, and S. epidermidis isolation was performed. The minimal inhibitory concentrations of 10 antibiotics were determined for the S. epidermidis isolates using the agar dilution method. High-level mupirocin-resistant isolates were subjected to whole-genome sequencing and bioinformatics analysis. A total of 94 un-duplicated S. epidermidis isolates were obtained from 107 facial skin samples. Results Antimicrobial susceptibility tests revealed that 23.4% of the 94 S. epidermidis isolates were resistant to oxacillin and positive for the mecA gene, which could be cauterized as methicillin-resistant S. epidermidis (MRSE). Resistance rates for erythromycin, clindamycin, tetracycline, ciprofloxacin, and gentamicin were 8.5%, 11.7%, 10.6%, 12.8%, and 1.1%, respectively. For mupirocin, the rates of low- and high-level resistance were 3.2% (3/94) and 11.7% (11/94), respectively. Resistance to vancomycin or linezolid was not observed. High-level mupirocin resistance in facial skin isolates is mediated by mupA. WGS and SNP-based phylogenetic analyses revealed diverse phylogenies among the 11 mupA-positive S. epidermidis isolates. Additionally, various resistance and virulence genes were identified in mupA-positive isolates. A new hybrid plasmid carrying mupA genes was found in two S. epidermidis isolates. Conclusion We observed a considerable level of antimicrobial resistance to several antibiotics and the prevalence of abundant and diverse resistance and virulence genes in the facial skin-origin S. epidermidis isolates. This may pose a potential risk for both public health and S. epidermidis infection.
Collapse
Affiliation(s)
- Bingqing Chen
- Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
| | - Lingyun Yao
- School of Perfume and Aroma Technology, Shanghai Institute of Technology, Shanghai, People’s Republic of China
| | - Rongjuan Cai
- School of Perfume and Aroma Technology, Shanghai Institute of Technology, Shanghai, People’s Republic of China
| | - Wei Chen
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, People’s Republic of China
| | - Yue Wang
- School of Perfume and Aroma Technology, Shanghai Institute of Technology, Shanghai, People’s Republic of China
- Institute of Shanghai Oriental Beauty Valley, Shanghai Institute of Technology, Shanghai, People’s Republic of China
| |
Collapse
|
42
|
Wang Y, Zhao M, Zou Y, Wang X, Zhang M, Sun Y. Hyaluronan Scaffold Decorated with Bifunctional Peptide Promotes Wound Healing via Antibacterial and Anti-Inflammatory. Biomacromolecules 2024; 25:7850-7860. [PMID: 39586057 DOI: 10.1021/acs.biomac.4c01130] [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: 11/27/2024]
Abstract
The invasion of bacteria and inflammation impeded infected wounds heal. Here, a hyaluronan-based scaffold (HAG-g-C) was designed by cross-linking with gallic acid-modified gelatin to provide a protein microenvironment and decorated with cathelicidin-BF (CBF), a natural antimicrobial peptide, to remove bacterial infections and reverse the inflammatory environment. In vitro, HAG-g-C presented an antibacterial effect on Staphylococcus aureus and Escherichia coli. Meanwhile, it could drive the phenotypic switch of macrophage from M1 to M2 to accelerate tissue remodeling. In a mouse model of S. aureus-infected total skin defects, HAG-g-C inhibited the process of infection at the beginning of the wound and then regulated the M1 macrophage transformed to M2 phenotype on day 12. In addition, HAG-g-C induced collagen deposition, and reduced the expression of TNF-α, thereby significantly accelerating the reconstruction of infected wounds.
Collapse
Affiliation(s)
- Yingzi Wang
- Electron Microscopy Laboratory of Renal Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P. R. China
| | - Mingda Zhao
- National Engineering Research Center for Biomaterials, Sichuan University, 29# Wangjiang Road, Chengdu, Sichuan 610064, P. R. China
- College of Biomedical Engineering, Sichuan University, 29# Wangjiang Road, Chengdu, Sichuan 610064, P. R. China
| | - Yaping Zou
- National Engineering Research Center for Biomaterials, Sichuan University, 29# Wangjiang Road, Chengdu, Sichuan 610064, P. R. China
- College of Biomedical Engineering, Sichuan University, 29# Wangjiang Road, Chengdu, Sichuan 610064, P. R. China
| | - Xiaojuan Wang
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Min Zhang
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Yong Sun
- National Engineering Research Center for Biomaterials, Sichuan University, 29# Wangjiang Road, Chengdu, Sichuan 610064, P. R. China
- College of Biomedical Engineering, Sichuan University, 29# Wangjiang Road, Chengdu, Sichuan 610064, P. R. China
| |
Collapse
|
43
|
Zheng Q, Li Y, Zhu W, Xu X, Sheng G, Li L. The Causal Effect Between Human Microbiota and Scabies: A Study from the Genetic Perspective. Clin Cosmet Investig Dermatol 2024; 17:2803-2812. [PMID: 39660030 PMCID: PMC11630723 DOI: 10.2147/ccid.s491660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Accepted: 11/29/2024] [Indexed: 12/12/2024]
Abstract
Background Previous studies have indicated that human flora may affect the development of scabies, however, no studies have proven a causal relationship between human flora and scabies, which would be detrimental to future in-depth studies on human flora and scabies. Methods Mendelian randomization (MR) was used to analyze the causal effect between human microbiota and scabies, with data on intestinal flora and skin flora from two large published studies and data on scabies from the FinnGen database. Five MR analysis methods were used to increase the reliability of the results, and sensitivity analyses were conducted to increase the robustness of the results. Results Our results suggest that 13 intestinal flora as well as 7 skin flora can have a causal effect on scabies. Conclusion Overall, our results demonstrate a causal relationship between intestinal and skin flora and scabies and are consistent with previous observational findings. This will contribute to the future development of probiotic agents for the prevention or treatment of scabies.
Collapse
Affiliation(s)
- Qi Zheng
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People’s Republic of China
| | - Yuetong Li
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People’s Republic of China
| | - Wenfeng Zhu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People’s Republic of China
| | - Xiang Xu
- Zhejiang Chinese Medical University, Hangzhou, People’s Republic of China
| | - Guoping Sheng
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Shulan (Hangzhou) Hospital Affiliated to Zhejiang Shuren University, Shulan International Medical College, Hangzhou, People’s Republic of China
| | - Lanjuan Li
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People’s Republic of China
| |
Collapse
|
44
|
Shan Z, Jiang B, Wang P, Wu W, Jin Y. Sustainable lignin-based composite hydrogels for controlled drug release and self-healing in antimicrobial wound dressing. Int J Biol Macromol 2024; 285:138327. [PMID: 39638185 DOI: 10.1016/j.ijbiomac.2024.138327] [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: 08/05/2024] [Revised: 11/14/2024] [Accepted: 12/02/2024] [Indexed: 12/07/2024]
Abstract
Bacterial infections pose a significant threat to global public health, demanding innovative solutions in biomedical field. Lignin is a naturally abundant polyphenol-rich polymer, offer promising potential to fabricate advance biomaterials for biomedical applications. Hence, a composite hydrogel with antimicrobial and antioxidant activities based on the development of dynamic covalent bonds among sodium alginate, lignin and epigallocatechin-3-gallate (EGCG) was designed. Lignin provides structural integrity to hydrogel backbone as well as released synergistically with the drug. This synergistic effect of the pH-responsive controlled release of both EGCG and lignin improved the releasing ability and bioactivity of the hydrogels. In in vitro antimicrobial experiments, the addition of 3.08 wt% lignin significantly enhanced bactericidal efficacy against Escherichia coli and Staphylococcus aureus, raising the killing rate from 20 % to over 96 %. The dynamic borate bond allows hydrogel network to repair itself when it is disrupted. Its self-healing ability, pH-responsive drug delivery, biocompatibility and strong antimicrobial and antioxidant effects make it a promising candidate for chronic wound management. This lignin-based hydrogel marks a significant innovation in sustainable, multifunctional biomedical materials.
Collapse
Affiliation(s)
- Zhu Shan
- Jiangsu Co-Innovation Center of Efficient Processing and Utilization of Forest Resources, International Innovation Center for Forest Chemicals and Materials, Nanjing Forestry University, Nanjing 210037, China; Jiangsu Provincial Key Lab of Pulp and Paper Science and Technology, College of Light Industry and Food Engineering, Nanjing Forestry University, Nanjing 210037, China
| | - Bo Jiang
- Jiangsu Co-Innovation Center of Efficient Processing and Utilization of Forest Resources, International Innovation Center for Forest Chemicals and Materials, Nanjing Forestry University, Nanjing 210037, China; Jiangsu Provincial Key Lab of Pulp and Paper Science and Technology, College of Light Industry and Food Engineering, Nanjing Forestry University, Nanjing 210037, China
| | - Peng Wang
- State Key Laboratory of Pharmaceutical Biotechnology, Department of Sports Medicine and Adult Reconstructive Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, China
| | - Wenjuan Wu
- Jiangsu Co-Innovation Center of Efficient Processing and Utilization of Forest Resources, International Innovation Center for Forest Chemicals and Materials, Nanjing Forestry University, Nanjing 210037, China; Jiangsu Provincial Key Lab of Pulp and Paper Science and Technology, College of Light Industry and Food Engineering, Nanjing Forestry University, Nanjing 210037, China
| | - Yongcan Jin
- Jiangsu Co-Innovation Center of Efficient Processing and Utilization of Forest Resources, International Innovation Center for Forest Chemicals and Materials, Nanjing Forestry University, Nanjing 210037, China; Jiangsu Provincial Key Lab of Pulp and Paper Science and Technology, College of Light Industry and Food Engineering, Nanjing Forestry University, Nanjing 210037, China.
| |
Collapse
|
45
|
Kohta M, Sugama J. Involvement of Urease-Producing Bacteria on Genital Skin in Community-Dwelling Women with Incontinence-Associated Dermatitis: A Cross-Sectional Study. J Multidiscip Healthc 2024; 17:5737-5747. [PMID: 39655293 PMCID: PMC11625642 DOI: 10.2147/jmdh.s492458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Accepted: 11/23/2024] [Indexed: 12/12/2024] Open
Abstract
Purpose Elevated skin pH facilitates the number of pathogenic bacteria increase, leading to the skin barrier dysfunction. This phenomenon is typically observed in individuals with Incontinence-associated dermatitis (IAD), which imposes a substantial physical and psychological burden on the afflicted individuals. We evaluated the association between the development of IAD in community-dwelling women with urinary incontinence and cutaneous urease-producing bacteria, as these bacteria may be involved in elevating skin pH by chemical reaction with urea in urine. Patients and Methods This was a cross-sectional study of 114 community-dwelling women with urinary incontinence who had registered for a survey campaign of a company. Swabs collected from genital skin were cultured in urea agar medium. The presence of urease-producing bacteria was determined by visually observing the change in the color of the culture medium caused by alkalization. The medium pH and total bacteria count were measured. Bacterial species were isolated and identified using a selective agar medium and simple identification kits. The participants were asked the presence of IAD by a self-administered questionnaire, and outcomes were compared between the IAD and no-IAD groups. This study followed the Strengthening the Reporting of Observational Studies in Epidemiology (STROBE) guideline. Results IAD was present in 31.6% (36/114) of participants. The detection rate of urease-producing bacteria and the medium pH was significantly higher in the IAD group than in the no-IAD group; however, the total number of bacteria was not significantly different. There were no significant between-group differences regarding the bacterial species identified. Conclusion The presence of urease-producing bacteria was associated with the development of IAD in community-dwelling women with urinary incontinence. Alkalization of the genital skin surface because of the contact between urine and urease-producing bacteria may compromise skin homeostasis. The bacterial species specifically involved in the development of IAD could not be determined.
Collapse
Affiliation(s)
- Masushi Kohta
- Research Center for Implementation Nursing Science Initiative, Fujita Health University, Toyoake, Aichi, Japan
| | - Junko Sugama
- Research Center for Implementation Nursing Science Initiative, Fujita Health University, Toyoake, Aichi, Japan
| |
Collapse
|
46
|
Kisielinski K, Wojtasik B, Zalewska A, Livermore DM, Jurczak-Kurek A. The bacterial burden of worn face masks-observational research and literature review. Front Public Health 2024; 12:1460981. [PMID: 39691656 PMCID: PMC11649673 DOI: 10.3389/fpubh.2024.1460981] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Accepted: 10/30/2024] [Indexed: 12/19/2024] Open
Abstract
Introduction Facemasks were widely mandated during the recent SARS-CoV-2 pandemic. Especially the use by the general population is associated with a higher risk of improper handling of the mask and contamination and potential adverse microbiological consequences. Methods We investigated and quantified bacterial accumulation in facemasks used by the general population, using 16S rRNA (Sanger Sequencing), culture and biochemical analysis along with Rose Bengal staining. Additionally, a systematic overview of the literature on face mask contamination was undertaken. Results We found an average bacterial load of 4.24 × 104 CFU recovered/mask, with a maximum load of 2.85 × 105 CFU. This maximum is 310 times higher than the limit value for contamination of ventilation system outlet surfaces specified by the German standard VDI 6022. Biochemical and molecular identification predominantly found Staphylococcus species (80%), including Staphylococcus aureus, along with endospore-forming Bacillus spp. Literature reports also indicate contamination of masks by bacterial and fungal opportunists of the genera Acinetobacter, Aspergillus, Alternaria, Bacillus, Cadosporium, Candida, Escherichia, Enterobacter, Enterococcus, Klebsiella (including K. pneumoniae), Micrococcus, Microsporum, Mucor, Pseudomonas, Staphylococcus and Streptococcus. Bacterial counts increase linearly with wearing duration. Discussion Prolonged use may affect the skin and respiratory microbiomes, promoting consequential eye, skin, oral and airway conditions. These aspects underscore the urgent need for further research and a risk-benefit analysis in respect of mask use, particularly given their unproven efficacy in disrupting the transmission of respiratory viruses and their adverse social consequences.
Collapse
Affiliation(s)
- Kai Kisielinski
- Clinical Medicine (Surgery), Emergency Medicine and Social Medicine, Private Practice, Düsseldorf, Germany
| | - Barbara Wojtasik
- Department of Evolutionary Genetics and Biosystematics, Faculty of Biology, University of Gdansk, Gdansk, Poland
| | - Aleksandra Zalewska
- Department of Evolutionary Genetics and Biosystematics, Faculty of Biology, University of Gdansk, Gdansk, Poland
| | - David M. Livermore
- Norwich Medical School, University of East Anglia, Norwich, United Kingdom
| | - Agata Jurczak-Kurek
- Department of Evolutionary Genetics and Biosystematics, Faculty of Biology, University of Gdansk, Gdansk, Poland
| |
Collapse
|
47
|
Zhang H, Zhu Y, Wang Y, Jiang L, Shi X, Cheng G. Microbial interactions shaping host attractiveness: insights into dynamic behavioral relationships. CURRENT OPINION IN INSECT SCIENCE 2024; 66:101275. [PMID: 39332621 DOI: 10.1016/j.cois.2024.101275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/16/2023] [Revised: 07/18/2024] [Accepted: 09/22/2024] [Indexed: 09/29/2024]
Abstract
Insects discern the presence of hosts (host plants) by integrating chemosensory, gustatory, and visual cues, with olfaction playing a pivotal role in this process. Among these factors, volatile signals produced by host-associated microbial communities significantly affect insect attraction. Microorganisms are widely and abundantly found on the surfaces of humans, plants, and insects. Notably, these microorganisms can metabolize compounds from the host surface and regulate the production of characteristic volatiles, which may guide the use of host microorganisms to modulate insect behavior. Essentially, the attraction of hosts to insects is intricately linked to the presence of their symbiotic microorganisms. This review underscores the critical role of microorganisms in shaping the dynamics of attractiveness between insects and their hosts.
Collapse
Affiliation(s)
- Hong Zhang
- New Cornerstone Science Laboratory, Tsinghua-Peking Joint Center for Life Sciences, School of Basic Medical Sciences, Tsinghua University, Beijing 100084, China; State Key Laboratory of Resource Insects, Institute of Apicultural Research, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Yibin Zhu
- New Cornerstone Science Laboratory, Tsinghua-Peking Joint Center for Life Sciences, School of Basic Medical Sciences, Tsinghua University, Beijing 100084, China; Institute of Infectious Diseases, Shenzhen Bay Laboratory, Shenzhen, Guangdong 518000, China; Institute of Pathogenic Organisms, Shenzhen Center for Disease Control and Prevention, Shenzhen 518055, China; Southwest United Graduate School, Kunming 650092, China
| | - Yibaina Wang
- China National Center for Food Safety Risk Assessment, Beijing 100022, China
| | - Liping Jiang
- Department of Parasitology, School of Basic Medical Sciences, Central South University, Changsha, Hunan 410013, China
| | - Xiaolu Shi
- Institute of Pathogenic Organisms, Shenzhen Center for Disease Control and Prevention, Shenzhen 518055, China
| | - Gong Cheng
- New Cornerstone Science Laboratory, Tsinghua-Peking Joint Center for Life Sciences, School of Basic Medical Sciences, Tsinghua University, Beijing 100084, China; Institute of Infectious Diseases, Shenzhen Bay Laboratory, Shenzhen, Guangdong 518000, China; Institute of Pathogenic Organisms, Shenzhen Center for Disease Control and Prevention, Shenzhen 518055, China; Southwest United Graduate School, Kunming 650092, China.
| |
Collapse
|
48
|
Wang Q, Wang M, Chen Y, Miao Q, Jin W, Ma Y, Pan J, Hu B. Deciphering microbiome and fungi-bacteria interactions in chronic wound infections using metagenomic sequencing. Eur J Clin Microbiol Infect Dis 2024; 43:2383-2396. [PMID: 39367927 DOI: 10.1007/s10096-024-04955-z] [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: 02/27/2024] [Accepted: 09/27/2024] [Indexed: 10/07/2024]
Abstract
PURPOSE Chronic wounds caused by infections impose a considerable global healthcare burden. The microbial features of these infections and possible correlations between bacteria and fungi may influence wound healing. However, metagenomic next-generation sequencing (mNGS) analyses of these features remain sparse. Therefore, we performed mNGS on chronic wound infection samples to investigate features and correlations between the bacteriome and mycobiome in 66 patients (28: chronic wounds; 38: non-chronic wounds). METHODS Microbial community characteristics in patients with wound infections, microbiome-systemic inflammation associations, and bacteria-fungi correlations were analyzed. RESULTS Infections constituted the primary cause of wounds in this study. Nontuberculous mycobacteria (23%) and Mycobacterium tuberculosis (13%) were the most common pathogens associated with chronic wounds, whereas Staphylococcus aureus (15%) was the most prevalent in non-chronic wound infections. Patients with chronic wound infections had a higher abundance of Pseudomonas aeruginosa than those without chronic wounds. Microbes with a high relative abundance in chronic wound infections were less significantly associated with plasma inflammatory factors than those in non-chronic wound infections. Additionally, a positive correlation between Candida glabrata and P. aeruginosa and an association between Malassezia restricta and anaerobic species were detected in patients with chronic wound infections. CONCLUSION Our results further support the hypothesis that P. aeruginosa is a microbial biomarker of chronic wound infection regardless of the causative pathogens. Moreover, we propose a positive correlation between C. glabrata and P. aeruginosa in chronic wound infections, which advances the current understanding of fungi-bacteria correlations in patients with chronic wound infections.
Collapse
Affiliation(s)
- Qingqing Wang
- Department of Infectious Diseases, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Meixia Wang
- Department of Hospital Infection Control, Zhongshan Hospital Xiamen Branch Hospital, Fudan University, Shanghai, China
| | - Yu Chen
- Department of Infectious Diseases, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Qing Miao
- Department of Infectious Diseases, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Wenting Jin
- Department of Infectious Diseases, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yuyan Ma
- Department of Infectious Diseases, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jue Pan
- Department of Infectious Diseases, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Bijie Hu
- Department of Infectious Diseases, Zhongshan Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
49
|
Kortekaas Krohn I, Callewaert C, Belasri H, De Pessemier B, Diez Lopez C, Mortz CG, O'Mahony L, Pérez-Gordo M, Sokolowska M, Unger Z, Untersmayr E, Homey B, Gomez-Casado C. The influence of lifestyle and environmental factors on host resilience through a homeostatic skin microbiota: An EAACI Task Force Report. Allergy 2024; 79:3269-3284. [PMID: 39485000 DOI: 10.1111/all.16378] [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: 01/28/2024] [Revised: 10/08/2024] [Accepted: 10/22/2024] [Indexed: 11/03/2024]
Abstract
Human skin is colonized with skin microbiota that includes commensal bacteria, fungi, arthropods, archaea and viruses. The composition of the microbiota varies at different anatomical locations according to changes in body temperature, pH, humidity/hydration or sebum content. A homeostatic skin microbiota is crucial to maintain epithelial barrier functions, to protect from invading pathogens and to interact with the immune system. Therefore, maintaining homeostasis holds promise to be an achievable goal for microbiome-directed treatment strategies as well as a prophylactic strategy to prevent the development of skin diseases, as dysbiosis or disruption of homeostatic skin microbiota is associated with skin inflammation. A healthy skin microbiome is likely modulated by genetic as well as environmental and lifestyle factors. In this review, we aim to provide a complete overview of the lifestyle and environmental factors that can contribute to maintaining the skin microbiome healthy. Awareness of these factors could be the basis for a prophylactic strategy to prevent the development of skin diseases or to be used as a therapeutic approach.
Collapse
Affiliation(s)
- Inge Kortekaas Krohn
- Vrije Universiteit Brussel (VUB), Skin Immunology & Immune Tolerance (SKIN) Research Group, Brussels, Belgium
- Vrije Universiteit Brussel (VUB), Department of Dermatology, Universitair Ziekenhuis Brussel (UZ Brussel), Brussels, Belgium
| | - Chris Callewaert
- Faculty of Bioscience Engineering, Ghent University, Centre for Microbial Ecology and Technology (CMET), Ghent, Belgium
| | - Hafsa Belasri
- Vrije Universiteit Brussel (VUB), Skin Immunology & Immune Tolerance (SKIN) Research Group, Brussels, Belgium
- Vrije Universiteit Brussel (VUB), Department of Dermatology, Universitair Ziekenhuis Brussel (UZ Brussel), Brussels, Belgium
| | - Britta De Pessemier
- Faculty of Bioscience Engineering, Ghent University, Centre for Microbial Ecology and Technology (CMET), Ghent, Belgium
| | - Celia Diez Lopez
- Faculty of Bioscience Engineering, Ghent University, Centre for Microbial Ecology and Technology (CMET), Ghent, Belgium
| | - Charlotte G Mortz
- Department of Dermatology and Allergy Centre, Odense University Hospital, University of Southern Denmark, Odense, Denmark
| | - Liam O'Mahony
- APC Microbiome Ireland, School of Microbiology, and Department of medicine, University College Cork, Cork, Ireland
| | - Marina Pérez-Gordo
- Departamento de Ciencias Médicas Básicas, Instituto de Medicina Molecular Aplicada (IMMA) Nemesio Díez, Facultad de Medicina, Universidad San Pablo-CEU, CEU Universities, Madrid, Spain
| | - Milena Sokolowska
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Zsofia Unger
- Department of Dermatology, University Hospital, Heinrich-Heine University, Duesseldorf, Germany
| | - Eva Untersmayr
- Institute of Pathophysiology and Allergy Research, Centre of Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Bernhard Homey
- Department of Dermatology, University Hospital, Heinrich-Heine University, Duesseldorf, Germany
| | - Cristina Gomez-Casado
- Department of Dermatology, University Hospital, Heinrich-Heine University, Duesseldorf, Germany
| |
Collapse
|
50
|
Madaan T, Doan K, Hartman A, Gherardini D, Ventrola A, Zhang Y, Kotagiri N. Advances in Microbiome-Based Therapeutics for Dermatological Disorders: Current Insights and Future Directions. Exp Dermatol 2024; 33:e70019. [PMID: 39641544 PMCID: PMC11663288 DOI: 10.1111/exd.70019] [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: 06/04/2024] [Revised: 10/30/2024] [Accepted: 11/19/2024] [Indexed: 12/07/2024]
Abstract
The human skin hosts an estimated 1000 bacterial species that are essential for maintaining skin health. Extensive clinical and preclinical studies have established the significant role of the skin microbiome in dermatological disorders such as atopic dermatitis, psoriasis, diabetic foot ulcers, hidradenitis suppurativa and skin cancers. In these conditions, the skin microbiome is not only altered but, in some cases, implicated in disease pathophysiology. Microbiome-based therapies (MBTs) represent an emerging category of live biotherapeutic products with tremendous potential as a novel intervention platform for skin diseases. Beyond using established wild-type strains native to the skin, these therapies can be enhanced to express targeted therapeutic molecules, offering more tailored treatment approaches. This review explores the role of the skin microbiome in various common skin disorders, with a particular focus on the development and therapeutic potential of MBTs for treating these conditions.
Collapse
Affiliation(s)
- Tushar Madaan
- James L. Winkle College of Pharmacy, University of Cincinnati, Cincinnati, OH 45267
| | - Kyla Doan
- James L. Winkle College of Pharmacy, University of Cincinnati, Cincinnati, OH 45267
| | - Alexandra Hartman
- James L. Winkle College of Pharmacy, University of Cincinnati, Cincinnati, OH 45267
| | - Dominick Gherardini
- James L. Winkle College of Pharmacy, University of Cincinnati, Cincinnati, OH 45267
| | - Alec Ventrola
- James L. Winkle College of Pharmacy, University of Cincinnati, Cincinnati, OH 45267
| | - Yuhang Zhang
- James L. Winkle College of Pharmacy, University of Cincinnati, Cincinnati, OH 45267
| | - Nalinikanth Kotagiri
- James L. Winkle College of Pharmacy, University of Cincinnati, Cincinnati, OH 45267
| |
Collapse
|