Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 10.463
Filtrer
1.
Sci Rep ; 14(1): 15188, 2024 07 02.
Article de Anglais | MEDLINE | ID: mdl-38956276

RÉSUMÉ

Wildlife harbour a diverse range of microorganisms that affect their health and development. Marsupials are born immunologically naïve and physiologically underdeveloped, with primary development occurring inside a pouch. Secretion of immunological compounds and antimicrobial peptides in the epithelial lining of the female's pouch, pouch young skin, and through the milk, are thought to boost the neonate's immune system and potentially alter the pouch skin microbiome. Here, using 16S rRNA amplicon sequencing, we characterised the Tasmanian devil pouch skin microbiome from 25 lactating and 30 non-lactating wild females to describe and compare across these reproductive stages. We found that the lactating pouch skin microbiome had significantly lower amplicon sequence variant richness and diversity than non-lactating pouches, however there was no overall dissimilarity in community structure between lactating and non-lactating pouches. The top five phyla were found to be consistent between both reproductive stages, with over 85% of the microbiome being comprised of Firmicutes, Proteobacteria, Fusobacteriota, Actinobacteriota, and Bacteroidota. The most abundant taxa remained consistent across all taxonomic ranks between lactating and non-lactating pouch types. This suggests that any potential immunological compounds or antimicrobial peptide secretions did not significantly influence the main community members. Of the more than 16,000 total identified amplicon sequence variants, 25 were recognised as differentially abundant between lactating and non-lactating pouches. It is proposed that the secretion of antimicrobial peptides in the pouch act to modulate these microbial communities. This study identifies candidate bacterial clades on which to test the activity of Tasmanian devil antimicrobial peptides and their role in pouch young protection, which in turn may lead to future therapeutic development for human diseases.


Sujet(s)
Lactation , Marsupialia , Microbiote , ARN ribosomique 16S , Animaux , Femelle , Marsupialia/microbiologie , ARN ribosomique 16S/génétique , Peau/microbiologie , Bactéries/classification , Bactéries/génétique
2.
Phytomedicine ; 130: 155542, 2024 Jul 25.
Article de Anglais | MEDLINE | ID: mdl-38823343

RÉSUMÉ

BACKGROUND: Malassezia globosa is a commensal basidiomycetous yeast occurring on the skin that causes pityriasis versicolor (PV) and seborrheic dermatitis, but that has also been implicated in other dermatoses. Cinnamaldehyde (CM) has antibacterial, antioxidant, and anti-inflammatory activities, but the effect of CM on M. globosa-infected PV has not been clarified. PURPOSE: The study aimed to investigate the possible antifungal and antibiofilm activities of CM against M. globosa-infected PV in vivo and in vitro. METHODS: The broth microdilution method was used to determine the minimum inhibitory concentration (MIC) of CM against M. globosa. The crystal violet staining assay and XTT assay were used to investigate the inhibition of CM on biofilm formation and the eradication of mature biofilms. The visualizations of the biofilm and cell distribution in the biofilm matrix were performed with a scanning electron microscope and confocal laser scanning microscope. The kits of antioxidant kinase were used to determine the activities of oxidative stress markers in M. globosa-stimulated HaCaT cells. Western blot assays were used to evaluate the role of TLR2/NF-κB in vitro. Furthermore, the protective effect of CM was assessed in M. globosa-associated PV mice. The expressions of inflammatory cytokines and apoptosis were screened using ELISA assays. The expressions of interleukin-6 and tumor necrosis factor-α were measured by an immunohistochemistry method in vivo. RESULTS: Our results showed that the MIC of CM against planktonic cells of M. globosa was 4 µg/ml and treatment with 20 × MIC CM eradicated mature biofilms of M. globosa. In vitro, after CM treatment the levels of oxidative stress indicators (i.e., superoxide dismutase, catalase, glutathione) significantly increased, while the levels of malondialdehyde decreased. In addition, the expression of TLR2/NF-κB in HaCaT cells was significantly reduced after CM treatment. On the other hand, an in vivo therapeutic effect of CM was assessed against M. globosa-infected mice. The fungal load on the skin decreased after treatment with CM compared to the M. globosa-infected group. In addition, the uninfected animals showed a normal skin structure, whereas, the M. globosa-infected mice showed extensive infiltration of neutrophils in skin tissues that improved after treatment with CM. Meanwhile, the levels of inflammatory and apoptotic factors improved after CM treatment. CONCLUSION: Our results showed that CM inhibits the biofilm formation of M. globosa and eradicates mature biofilms of M. globosa. Treatment with CM significantly decreased oxidative stress, apoptosis, and inflammatory markers in the skin tissue and HaCaT cells. Hence, this study suggests that CM is a good candidate therapeutic agent against M. globosa-induced PV infections because of its antifungal, antibiofilm, and anti-inflammatory properties.


Sujet(s)
Acroléine , Antifongiques , Biofilms , Malassezia , Tests de sensibilité microbienne , Pityriasis versicolor , Récepteur de type Toll-2 , Biofilms/effets des médicaments et des substances chimiques , Acroléine/analogues et dérivés , Acroléine/pharmacologie , Animaux , Malassezia/effets des médicaments et des substances chimiques , Humains , Récepteur de type Toll-2/métabolisme , Pityriasis versicolor/traitement médicamenteux , Antifongiques/pharmacologie , Souris , Stress oxydatif/effets des médicaments et des substances chimiques , Cellules HaCaT , Facteur de transcription NF-kappa B/métabolisme , Interleukine-6/métabolisme , Antioxydants/pharmacologie , Facteur de nécrose tumorale alpha/métabolisme , Peau/effets des médicaments et des substances chimiques , Peau/microbiologie
3.
Arch Dermatol Res ; 316(7): 374, 2024 Jun 08.
Article de Anglais | MEDLINE | ID: mdl-38850443

RÉSUMÉ

The microbiome is intricately linked to the development of psoriasis, serving as both a potential cause and consequence of the psoriatic process. In recent years, there has been growing interest among psoriasis researchers in exploring how psoriasis treatments affect the skin and gut microbiome. However, a comprehensive evaluation of the impact of modern treatment approaches on the microbiome has yet to be conducted. In this systematic review, we analyze studies investigating alterations in the skin and gut microbiome resulting from psoriasis treatment, aiming to understand how current therapies influence the role of the microbiome in psoriasis development. The systematic review was performed according to the Preferred Reporting Items for Systematic Reviews and Meta-Analysis (PRISMA) guidelines. PubMed and Scopus databases were searched for eligible studies from the inception dates until July 5, 2023. Study selection, data extraction, and risk of bias assessment were carried out by three overlapping pairs of reviewers, resolving any disagreements through consensus. Our analysis of various treatments, including biologics, conventional medications, phototherapy, and probiotics, reveals significant shifts in microbial diversity and abundance. Importantly, favorable treatment outcomes are associated with microbiota alterations that approach those observed in healthy individuals. While the studies reviewed exhibit varying degrees of bias, underscoring the need for further research, this review supports the potential of microbiome modulation as both a preventive and therapeutic strategy for psoriasis patients. The findings underscore the importance of personalized therapeutic approaches, recognizing the profound impact of treatment on the microbiome. They also highlight the promise of probiotics, prebiotics, and dietary interventions in psoriasis management.


Sujet(s)
Microbiome gastro-intestinal , Probiotiques , Psoriasis , Peau , Psoriasis/microbiologie , Psoriasis/immunologie , Psoriasis/thérapie , Humains , Microbiome gastro-intestinal/effets des médicaments et des substances chimiques , Microbiome gastro-intestinal/physiologie , Peau/microbiologie , Probiotiques/administration et posologie , Photothérapie/méthodes , Produits biologiques/usage thérapeutique , Résultat thérapeutique , Produits dermatologiques/usage thérapeutique , Produits dermatologiques/administration et posologie
4.
Pol J Microbiol ; 73(2): 199-206, 2024 Jun 01.
Article de Anglais | MEDLINE | ID: mdl-38905277

RÉSUMÉ

Negative Pressure Wound Therapy (NPWT) has been widely adopted in wound healing strategies due to its multimodal mechanism of action. While NPWT's positive impression on wound healing is well-established, its effect on bacterial load reduction remains equivocal. This study investigates NPWT's efficacy in reducing bioburden using an in vitro porcine skin model, focusing on the impact of Staphylococcus aureus and Staphylococcus epidermidis. Custom-made negative pressure chambers were employed to apply varying negative pressures. Porcine skin was cut into 5 × 5 cm squares and three standardized wounds of 6 mm each were created using a biopsy punch. Then, wounds were infected with S. aureus and S. epidermidis bacterial suspensions diluted 1:10,000 to obtain a final concentration of 1.5 × 104 CFU/ml and were placed in negative pressure chambers. After incubation, bacterial counts were expressed as colony-forming units (CFU) per ml. For S. aureus at 120 hours, the median CFU, mean area per colony, and total growth area were notably lower at -80 mmHg when compared to -250 mmHg and -50 mmHg, suggesting an optimal negative pressure for the pressure-dependent inhibition of the bacterial proliferation. While analyzing S. epidermidis at 120 hours, the response to the negative pressure was similar but less clear, with the minor CFU at -100 mmHg. The influence of intermittent negative pressure on the S. epidermidis growth showed notably lower median CFU with the interval therapy every hour compared to the S. aureus control group. This study contributes valuable insights into NPWT's influence on the bacterial load, emphasizing the need for further research to reformulate its role in managing contaminated wounds.


Sujet(s)
Traitement des plaies par pression négative , Staphylococcus aureus , Staphylococcus epidermidis , Staphylococcus epidermidis/croissance et développement , Staphylococcus epidermidis/physiologie , Animaux , Suidae , Staphylococcus aureus/croissance et développement , Staphylococcus aureus/physiologie , Cicatrisation de plaie , Charge bactérienne , Infection de plaie/microbiologie , Infection de plaie/thérapie , Cinétique , Infections à staphylocoques/microbiologie , Peau/microbiologie
5.
Int J Mycobacteriol ; 13(2): 218-220, 2024 Apr 01.
Article de Anglais | MEDLINE | ID: mdl-38916395

RÉSUMÉ

Leprosy, caused by the bacterium Mycobacterium leprae, is known to primarily affect the skin and peripheral nerves. We present a rare case of leprosy initially manifesting as demyelinating polyneuropathy. A 46-year-old female presented with progressive weakness, tingling, and numbness in her extremities. Nerve conduction studies revealed evidence of demyelination, prompting further investigations. Skin slit-skin smears confirmed the diagnosis of leprosy, with the presence of acid-fast bacilli. The patient was subsequently started on multidrug therapy, leading to significant clinical improvement. This case highlights the importance of considering leprosy as a differential diagnosis in patients presenting with demyelinating polyneuropathy, especially in endemic regions.


Sujet(s)
Lèpre , Mycobacterium leprae , Polyradiculonévrite inflammatoire démyélinisante chronique , Humains , Femelle , Adulte d'âge moyen , Polyradiculonévrite inflammatoire démyélinisante chronique/diagnostic , Polyradiculonévrite inflammatoire démyélinisante chronique/traitement médicamenteux , Lèpre/diagnostic , Lèpre/traitement médicamenteux , Lèpre/microbiologie , Lèpre/complications , Diagnostic différentiel , Mycobacterium leprae/isolement et purification , Mycobacterium leprae/génétique , Peau/anatomopathologie , Peau/microbiologie , Antilépreux/usage thérapeutique
6.
Front Immunol ; 15: 1354154, 2024.
Article de Anglais | MEDLINE | ID: mdl-38903509

RÉSUMÉ

Background: Atopic dermatitis (AD) is a common chronic inflammatory skin diseases that seriously affects life quality of the patients. Staphylococcus aureus (S. aureus) colonization on the skin plays an important role in the pathogenesis of AD; however, the mechanism of how it modulates skin immunity to exacerbate AD remains unclear. MicroRNAs are short non-coding RNAs that act as post-transcriptional regulators of genes. They are involved in the pathogenesis of various inflammatory skin diseases. Methods: In this study, we established miRNA expression profiles for keratinocytes stimulated with heat-killed S. aureus (HKSA). The expression of miR-939 in atopic dermatitis patients was analyzed by fluorescence in situ hybridization (FISH). miR-939 mimic was transfected to human primary keratinocyte to investigate its impact on the expression of matrix metalloproteinase genes (MMPs) in vitro. Subsequently, miR-939, along with Polyplus transfection reagent, was administered to MC903-induced atopic dermatitis skin to assess its function in vivo. Results: MiR-939 was highly upregulated in HKSA-stimulated keratinocytes and AD lesions. In vitro studies revealed that miR-939 increased the expression of matrix metalloproteinase genes, including MMP1, MMP3, and MMP9, as well as the cell adhesion molecule ICAM1 in human primary keratinocytes. In vivo studies indicated that miR-939 increased the expression of matrix metalloproteinases to promote the colonization of S. aureus and exacerbated S. aureus-induced AD-like skin inflammation. Conclusions: Our work reveals miR-939 is an important regulator of skin inflammation in AD that could be used as a potential therapeutic target for AD.


Sujet(s)
Eczéma atopique , Kératinocytes , Matrix metalloproteinases , microARN , Staphylococcus aureus , Eczéma atopique/immunologie , Eczéma atopique/génétique , Humains , microARN/génétique , Kératinocytes/métabolisme , Kératinocytes/immunologie , Animaux , Souris , Matrix metalloproteinases/génétique , Matrix metalloproteinases/métabolisme , Infections à staphylocoques/immunologie , Infections à staphylocoques/génétique , Femelle , Mâle , Modèles animaux de maladie humaine , Peau/microbiologie , Peau/anatomopathologie , Peau/immunologie , Cellules cultivées
7.
NPJ Biofilms Microbiomes ; 10(1): 50, 2024 Jun 20.
Article de Anglais | MEDLINE | ID: mdl-38902263

RÉSUMÉ

During the COVID-19 pandemic, facemasks played a pivotal role in preventing person-person droplet transmission of viral particles. However, prolonged facemask wearing causes skin irritations colloquially referred to as 'maskne' (mask + acne), which manifests as acne and contact dermatitis and is mostly caused by pathogenic skin microbes. Previous studies revealed that the putative causal microbes were anaerobic bacteria, but the pathogenesis of facemask-associated skin conditions remains poorly defined. We therefore characterized the role of the facemask-associated skin microbiota in the development of maskne using culture-dependent and -independent methodologies. Metagenomic analysis revealed that the majority of the facemask microbiota were anaerobic bacteria that originated from the skin rather than saliva. Previous work demonstrated direct interaction between pathogenic bacteria and antagonistic strains in the microbiome. We expanded this analysis to include indirect interaction between pathogenic bacteria and other indigenous bacteria classified as either 'pathogen helper (PH)' or 'pathogen inhibitor (PIn)' strains. In vitro screening of bacteria isolated from facemasks identified both strains that antagonized and promoted pathogen growth. These data were validated using a mouse skin infection model, where we observed attenuation of symptoms following pathogen infection. Moreover, the inhibitor of pathogen helper (IPH) strain, which did not directly attenuate pathogen growth in vitro and in vivo, functioned to suppress symptom development and pathogen growth indirectly through PH inhibitory antibacterial products such as phenyl lactic acid. Taken together, our study is the first to define a mechanism by which indirect microbiota interactions under facemasks can control symptoms of maskne by suppressing a skin pathogen.


Sujet(s)
COVID-19 , Masques , Microbiote , Peau , Animaux , Souris , Humains , COVID-19/microbiologie , COVID-19/virologie , Peau/microbiologie , Acné juvénile/microbiologie , SARS-CoV-2 , Femelle , Métagénomique/méthodes , Modèles animaux de maladie humaine , Bactéries/classification , Bactéries/génétique , Bactéries/isolement et purification , Interactions microbiennes , Eczéma de contact/étiologie
8.
Nat Commun ; 15(1): 4725, 2024 Jun 03.
Article de Anglais | MEDLINE | ID: mdl-38830879

RÉSUMÉ

Non-genetic sources of phenotypic variation, such as the epigenome and the microbiome, could be important contributors to adaptive variation for species with low genetic diversity. However, little is known about the complex interaction between these factors and the genetic diversity of the host, particularly in wild populations. Here, we examine the skin microbiome composition of two closely-related mangrove killifish species with different mating systems (self-fertilising and outcrossing) under sympatric and allopatric conditions. This allows us to partition the influence of the genotype and the environment on their microbiome and (previously described) epigenetic profiles. We find the diversity and community composition of the skin microbiome are strongly shaped by the environment and, to a lesser extent, by species-specific influences. Heterozygosity and microbiome alpha diversity, but not epigenetic variation, are associated with the fluctuating asymmetry of traits related to performance (vision) and behaviour (aggression). Our study identifies that a proportion of the epigenetic diversity and microbiome differentiation is unrelated to genetic variation, and we find evidence for an associative relationship between microbiome and epigenetic diversity in these wild populations. This suggests that both mechanisms could potentially contribute to variation in species with low genetic diversity.


Sujet(s)
Épigenèse génétique , Variation génétique , Microbiote , Animaux , Microbiote/génétique , Peau/microbiologie , Cyprinodontiformes/génétique , Cyprinodontiformes/microbiologie , Mâle , Génotype , Spécificité d'espèce , Femelle
9.
J Oleo Sci ; 73(7): 953-961, 2024.
Article de Anglais | MEDLINE | ID: mdl-38945924

RÉSUMÉ

Handwashing represents an important personal hygiene measure for preventing infection. Herein, we report the persistence of antibacterial and antiviral effects after handwashing with fatty acid salt-based hand soap. To this end, we developed a new in vitro test method to measure persistence, utilizing coacervation formed by anionic surfactants and cationic polymers to retain highly effective soap components against each bacterium and virus on the skin. Coacervation with fatty acid salts and poly diallyldimethylammonium chloride (PDADMAC) as a cationic polymer allowed the persistence of antibacterial and antiviral effects against E. coli, S. aureus, and influenza virus even 4 h after handwashing. Furthermore, we confirmed an increase in the number of residual components effective against each bacterium and virus on the skin. In summary, the current findings describe an effective approach for enhancing the protective effects of handwashing.


Sujet(s)
Antibactériens , Antiviraux , Escherichia coli , Désinfection des mains , Polyéthylènes , Composés d'ammonium quaternaire , Peau , Savons , Staphylococcus aureus , Tensioactifs , Savons/pharmacologie , Escherichia coli/effets des médicaments et des substances chimiques , Désinfection des mains/méthodes , Composés d'ammonium quaternaire/pharmacologie , Antibactériens/pharmacologie , Staphylococcus aureus/effets des médicaments et des substances chimiques , Antiviraux/pharmacologie , Peau/effets des médicaments et des substances chimiques , Peau/microbiologie , Tensioactifs/pharmacologie , Humains , Acides gras/pharmacologie , Acides gras/analyse , Facteurs temps , Orthomyxoviridae/effets des médicaments et des substances chimiques
11.
Biosens Bioelectron ; 261: 116467, 2024 Oct 01.
Article de Anglais | MEDLINE | ID: mdl-38901392

RÉSUMÉ

Light therapy is an effective approach for the treatment of a variety of challenging dermatological conditions. In contrast to existing methods involving high doses and large areas of illumination, alternative strategies based on wearable designs that utilize a low light dose over an extended period provide a precise and convenient treatment. In this study, we present a battery-free, skin-integrated optoelectronic patch that incorporates a coil-powered circuit, an array of microscale violet and red light emitting diodes (LEDs), and polymer microneedles (MNs) loaded with 5-aminolevulinic acid (5-ALA). These polymer MNs, based on the biodegradable composite materials of polyvinyl alcohol (PVA) and hyaluronic acid (HA), serve as light waveguides for optical access and a medium for drug release into deeper skin layers. Unlike conventional clinical photomedical appliances with a rigid and fixed light source, this flexible design allows for a conformable light source that can be applied directly to the skin. In animal models with bacterial-infected wounds, the experimental group with the combination treatment of metronomic photodynamic and light therapies reduced 2.48 log10 CFU mL-1 in bactericidal level compared to the control group, indicating an effective anti-infective response. Furthermore, post-treatment analysis revealed the activation of proregenerative genes in monocyte and macrophage cell populations, suggesting enhanced tissue regeneration, neovascularization, and dermal recovery. Overall, this optoelectronic patch design broadens the scope for targeting deep skin lesions, and provides an alternative with the functionality of standard clinical light therapy methods.


Sujet(s)
Photothérapie dynamique , Animaux , Photothérapie dynamique/méthodes , Souris , Humains , Poly(alcool vinylique)/composition chimique , Acide amino-lévulinique/usage thérapeutique , Acide amino-lévulinique/pharmacologie , Acide amino-lévulinique/composition chimique , Acide amino-lévulinique/administration et posologie , Techniques de biocapteur , Acide hyaluronique/composition chimique , Infection de plaie/traitement médicamenteux , Infection de plaie/microbiologie , Infection de plaie/thérapie , Photosensibilisants/composition chimique , Photosensibilisants/pharmacologie , Peau/effets des radiations , Peau/microbiologie , Conception d'appareillage
12.
Acta Derm Venereol ; 104: adv34892, 2024 Jun 19.
Article de Anglais | MEDLINE | ID: mdl-38898675

RÉSUMÉ

Psoriasis results from both genetic predisposition and environmental triggers, such as Streptococcal infections. This study aimed to explore the correlation between the abundance of the Streptococcus genus on the skin and psoriasis severity in individuals carrying specific psoriasis-associated genetic variants. Studying 39 chronic plaque psoriasis patients, the elbow skin microbiome and 49 psoriasis-related single nucleotide polymorphisms (SNPs) were analysed using a MiSeq instrument for 16S rDNA sequencing, and CLC Genomic Workbench for processing and analysis. Through multivariate linear regression analysis, a positive correlation was found between Streptococcus genus abundance and psoriasis severity in patients with certain FBXL19 gene-related heterozygous SNPs (rs12924903, rs10782001, rs12445568). Conversely, a negative association was observed in patients with homozygous genotypes. Moreover, we identified an association between Streptococcus abundance and psoriasis severity in patients with genetic variants related to IL-22, ERAP1, NOS2, and ILF3. This is the first study highlighting a positive association between Streptococcus skin colonization and psoriasis severity in patients with heterozygous genotypes within the FBXL19 gene region. FXBL19 targets the IL-33/IL1RL1 axis, crucial in infectious diseases and innate immunity promotion. These novel results suggests an intricate interaction among host genetics, Streptococcus skin colonization, and psoriasis inflammation, offering potential avenues for novel treatment approaches.


Sujet(s)
Protéines F-box , Polymorphisme de nucléotide simple , Psoriasis , Indice de gravité de la maladie , Peau , Streptococcus , Humains , Mâle , Psoriasis/génétique , Psoriasis/microbiologie , Femelle , Adulte d'âge moyen , Adulte , Peau/microbiologie , Streptococcus/génétique , Streptococcus/isolement et purification , Protéines F-box/génétique , Prédisposition génétique à une maladie , Phénotype , Hétérozygote , Interactions hôte-pathogène , Homozygote , Ribotypage , Sujet âgé
13.
Ren Fail ; 46(2): 2363417, 2024 Dec.
Article de Anglais | MEDLINE | ID: mdl-38913582

RÉSUMÉ

OBJECTIVES: Hemodialysis patients with end-stage renal disease (ESRD) are susceptible to infections and dysbiosis. Catheter-related infections are typically caused by opportunistic skin pathogens. This study aims to compare the skin microbiota changes around the exit site of tunneled cuffed catheters (peri-catheter group) and the contralateral site (control group). METHODS: ESRD patients on hemodialysis were recruited. The skin microbiota were collected with moist skin swabs and analyzed using high-throughput sequencing of the 16S rDNA V3-V4 region. After denoising, de-replication, and removal of chimeras, the reads were assigned to zero-radius operational taxonomic units (ZOTU). RESULTS: We found significantly reduced alpha diversity in the peri-catheter group compared to the control group, as indicated by the Shannon, Jost, and equitability indexes, but not by the Chao1 or richness indexes. Beta diversity analysis revealed significant deviation of the peri-catheter microbiota from its corresponding control group. There was an overrepresentation of Firmicutes and an underrepresentation of Actinobacteria, Proteobacteria, and Acidobacteria at the phylum level in the peri-catheter group. The most abundant ZOTU (Staphylococcus spp.) drastically increased, while Cutibacterium, a commensal bacterium, decreased in the peri-catheter group. Network analysis revealed that the skin microbiota demonstrated covariance with both local and biochemical factors. CONCLUSIONS: In conclusion, there was significant skin microbiota dysbiosis at the exit sites compared to the control sites in ESRD dialysis patients. Managing skin dysbiosis represents a promising target in the prevention of catheter-related bacterial infections.


Sujet(s)
Dysbiose , Défaillance rénale chronique , Microbiote , Dialyse rénale , Peau , Staphylococcus , Humains , Adulte d'âge moyen , Mâle , Dialyse rénale/effets indésirables , Dialyse rénale/instrumentation , Femelle , Peau/microbiologie , Défaillance rénale chronique/thérapie , Défaillance rénale chronique/complications , Dysbiose/microbiologie , Dysbiose/étiologie , Sujet âgé , Staphylococcus/isolement et purification , Infections sur cathéters/microbiologie , Voies veineuses centrales/effets indésirables , Voies veineuses centrales/microbiologie , Adulte , ARN ribosomique 16S/génétique
14.
NPJ Biofilms Microbiomes ; 10(1): 52, 2024 Jun 25.
Article de Anglais | MEDLINE | ID: mdl-38918415

RÉSUMÉ

It is becoming increasingly apparent that commensal skin bacteria have an important role in wound healing and infection progression. However, the precise mechanisms underpinning many of these probiotic interactions remain to be fully uncovered. In this work, we demonstrate that the common skin commensal Cutibacterium acnes can limit the pathogenicity of the prevalent wound pathogen Pseudomonas aeruginosa in vivo. We show that this impact on pathogenicity is independent of any effect on growth, but occurs through a significant downregulation of the Type Three Secretion System (T3SS), the primary toxin secretion system utilised by P. aeruginosa in eukaryotic infection. We also show a downregulation in glucose acquisition systems, a known regulator of the T3SS, suggesting that glucose availability in a wound can influence infection progression. C. acnes is well known as a glucose fermenting organism, and we demonstrate that topically supplementing a wound with glucose reverses the probiotic effects of C. acnes. This suggests that introducing carbon source competition within the wound microenvironment may be an effective way to prevent or limit wound infection.


Sujet(s)
Glucose , Pseudomonas aeruginosa , Pseudomonas aeruginosa/métabolisme , Pseudomonas aeruginosa/pathogénicité , Glucose/métabolisme , Animaux , Systèmes de sécrétion de type III/métabolisme , Systèmes de sécrétion de type III/génétique , Propionibacterium acnes/croissance et développement , Propionibacterium acnes/physiologie , Propionibacterium acnes/métabolisme , Infection de plaie/microbiologie , Souris , Infections à Pseudomonas/microbiologie , Peau/microbiologie , Carbone/métabolisme , Cicatrisation de plaie , Antibiose , Évolution de la maladie , Humains
15.
Int J Mol Sci ; 25(12)2024 Jun 13.
Article de Anglais | MEDLINE | ID: mdl-38928245

RÉSUMÉ

Atopic dermatitis (AD) is a chronic inflammatory skin condition with a high prevalence worldwide. AD pathogenesis is complex and consists of immune system dysregulation and impaired skin barrier, influenced by genetic and environmental factors. The purpose of the review is to show the complex interplay between atopic dermatitis and the microbiota. Human microbiota plays an important role in AD pathogenesis and the course of the disease. Dysbiosis is an important factor contributing to the development of atopic diseases, including atopic dermatitis. The gut microbiota can influence the composition of the skin microbiota, strengthening the skin barrier and regulating the immune response via the involvement of bacterial metabolites, particularly short-chain fatty acids, in signaling pathways of the gut-skin axis. AD can be modulated by antibiotic intake, dietary adjustments, hygiene, and living conditions. One of the promising strategies for modulating the course of AD is probiotics. This review offers a summary of how the microbiota influences the development and treatment of AD, highlighting aspects that warrant additional investigation.


Sujet(s)
Eczéma atopique , Dysbiose , Microbiome gastro-intestinal , Probiotiques , Eczéma atopique/microbiologie , Eczéma atopique/thérapie , Humains , Dysbiose/microbiologie , Dysbiose/thérapie , Probiotiques/usage thérapeutique , Microbiote , Peau/microbiologie , Animaux
16.
AAPS PharmSciTech ; 25(5): 130, 2024 Jun 06.
Article de Anglais | MEDLINE | ID: mdl-38844611

RÉSUMÉ

Naringenin (NRG) inhibits the fungal 17ß-hydroxysteroid dehydrogenase accountable for ergosterol synthesis in Candida albicans (C. albicans), a causative agent for cutaneous candidiasis. In present research, NRG was complexed with ZnO nanomaterial (NRG-Zn2+) to synthesize NRG-Zn2+ nanocomposites. The particle size and ζ-potential of NRG-Zn2+ nanocomposites were respectively estimated to be 180.33 ± 1.22-nm and - 3.92 ± 0.35-mV. In silico data predicted the greater affinity of NRG-Zn2+ nanocomposite for 14α-demethylase and ceramide in comparison to NRG alone. Later, NRG-Zn2+ nanocomposites solution was transformed in to naringenin-zinc oxide nanocomposites loaded chitosan gel (NRG-Zn-CS-Gel) with viscosity and firmness of 854806.7 ± 52386.43 cP and 698.27 ± 10.35 g, respectively. The ex-vivo skin permeation demonstrated 70.49 ± 5.22% skin retention, significantly greater (P < 0.05) than 44.48 ± 3.06% of naringenin loaded chitosan gel (NRG-CS-Gel) and 31.24 ± 3.28% of naringenin solution (NRG Solution). NRG-Zn-CS-Gel demonstrated 6.71 ± 0.84% permeation of NRG with a flux value of 0.046 ± 0.01-µg/cm2/h. The MIC50 of NRG-Zn-CS-Gel against C. albicans was estimated to be 0.156-µg/mL with FICI (fractional inhibitory concentration index) of 0.018 that consequently exhibited synergistic efficacy. Further, NRG-Zn-CS-Gel demonstrated superior antifungal efficacy in C. albicans induced cutaneous candidiasis infection in Balb/c mice. The fungal burden in NRG-Zn-CS-Gel treated group was 109 ± 25 CFU/mL, significantly lower (P < 0.05) than positive control (2260 ± 446 CFU/mL), naringenin loaded chitosan gel (NRG-CS-Gel; 928 ± 127 CFU/mL) and chitosan gel (CS-Gel; 2116 ± 186 CFU/mL) treated mice. Further, histopathology examination and cytokine profiling of TNF-α, IL-1ß and IL-10 revealed the healing of skin and inflammation associated with cutaneous candidiasis infection. In conclusion, NRG-Zn-CS-Gel may be a potential candidate for translating in to a clinical viable topical nanotherapeutic.


Sujet(s)
Antifongiques , Candida albicans , Chitosane , Flavanones , Gels , Souris de lignée BALB C , Nanocomposites , Oxyde de zinc , Animaux , Flavanones/administration et posologie , Flavanones/pharmacologie , Souris , Candida albicans/effets des médicaments et des substances chimiques , Chitosane/composition chimique , Chitosane/administration et posologie , Nanocomposites/composition chimique , Nanocomposites/administration et posologie , Antifongiques/administration et posologie , Antifongiques/pharmacologie , Antifongiques/pharmacocinétique , Oxyde de zinc/administration et posologie , Oxyde de zinc/pharmacologie , Oxyde de zinc/composition chimique , Systèmes de délivrance de médicaments/méthodes , Peau/métabolisme , Peau/effets des médicaments et des substances chimiques , Peau/microbiologie , Candidose/traitement médicamenteux , Polymères/composition chimique , Absorption cutanée/effets des médicaments et des substances chimiques , Taille de particule , Administration par voie cutanée
17.
Acta Derm Venereol ; 104: adv34882, 2024 Jun 11.
Article de Anglais | MEDLINE | ID: mdl-38860624

RÉSUMÉ

Patients with atopic dermatitis (AD) are more likely than healthy individuals to harbour Staphylococcus aureus on their skin. Superantigens (SAgs) produced by specific S. aureus strains may contribute to AD-associated skin inflammation. The present study compared the prevalence and types of SAg-encoding genes between S. aureus isolated from patients with AD and from  controls, and within the AD group between isolates from different sampling sites (lesional skin, non-lesional skin, and nares). This retrospective case-control study extracted data from 2 previous studies that examined S. aureus using whole-genome sequencing. The 138 S. aureus isolates obtained from 71 AD patients contained 349 SAg-encoding genes; 22 (6.3%) were found in isolates from nares (0.4 ± 0.6 genes per isolate), 99 (28.4%) in isolates from non-lesional skin (3.7 ± 3.9), and 228 (65.3%) in isolates from lesional skin (4.2 ± 4.5). S. aureus (n = 101) from the control group contained 594 SAg-encoding genes (5.9 ± 4.2). Of the S. aureus isolated from lesional AD skin, 69% carried at least 1 gene encoding SAg compared with 33% of AD nasal isolates. SAg could be a factor in the pathogenesis of a subset of AD patients.


Sujet(s)
Eczéma atopique , Peau , Staphylococcus aureus , Superantigènes , Humains , Eczéma atopique/microbiologie , Superantigènes/génétique , Staphylococcus aureus/génétique , Staphylococcus aureus/isolement et purification , Études rétrospectives , Peau/microbiologie , Mâle , Femelle , Études cas-témoins , Adulte , Infections cutanées à staphylocoques/microbiologie , Adulte d'âge moyen , Jeune adulte
18.
ISME J ; 18(1)2024 Jan 08.
Article de Anglais | MEDLINE | ID: mdl-38861457

RÉSUMÉ

Phylosymbiosis is an association between host-associated microbiome composition and host phylogeny. This pattern can arise via the evolution of host traits, habitat preferences, diets, and the co-diversification of hosts and microbes. Understanding the drivers of phylosymbiosis is vital for modelling disease-microbiome interactions and manipulating microbiomes in multi-host systems. This study quantifies phylosymbiosis in Appalachian salamander skin in the context of infection by the fungal pathogen Batrachochytrium dendrobatidis (Bd), while accounting for environmental microbiome exposure. We sampled ten salamander species representing >150M years of divergence, assessed their Bd infection status, and analysed their skin and environmental microbiomes. Our results reveal a significant signal of phylosymbiosis, whereas the local environmental pool of microbes, climate, geography, and Bd infection load had a smaller impact. Host-microbe co-speciation was not evident, indicating that the effect stems from the evolution of host traits influencing microbiome assembly. Bd infection is correlated with host phylogeny and the abundance of Bd-inhibitory bacterial strains, suggesting that the long-term evolutionary dynamics between salamander hosts and their skin microbiomes affect the present-day distribution of the pathogen, along with habitat-linked exposure risk. Five Bd-inhibitory bacterial strains showed unusual generalism: occurring in most host species and habitats. These generalist strains may enhance the likelihood of probiotic manipulations colonising and persisting on hosts. Our results underscore the substantial influence of host-microbiome eco-evolutionary dynamics on environmental health and disease outcomes.


Sujet(s)
Batrachochytrium (genre) , Microbiote , Phylogenèse , Peau , Symbiose , Urodela , Animaux , Urodela/microbiologie , Peau/microbiologie , Batrachochytrium (genre)/génétique , Bactéries/classification , Bactéries/génétique , Bactéries/isolement et purification
19.
Mycoses ; 67(6): e13753, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38877612

RÉSUMÉ

Candida species, commensal residents of human skin, are recognized as the cause of cutaneous candidiasis across various body surfaces. Individuals with weakened immune systems, particularly those with immunosuppressive conditions, are significantly more susceptible to this infection. Diabetes mellitus, a major metabolic disorder, has emerged as a critical factor inducing immunosuppression, thereby facilitating Candida colonization and subsequent skin infections. This comprehensive review examines the prevalence of different types of Candida albicans-induced cutaneous candidiasis in diabetic patients. It explores the underlying mechanisms of pathogenicity and offers insights into recommended preventive measures and treatment strategies. Diabetes notably increases vulnerability to oral and oesophageal candidiasis. Additionally, it can precipitate vulvovaginal candidiasis in females, Candida balanitis in males, and diaper candidiasis in young children with diabetes. Diabetic individuals may also experience candidal infections on their nails, hands and feet. Notably, diabetes appears to be a risk factor for intertrigo syndrome in obese individuals and periodontal disorders in denture wearers. In conclusion, the intricate relationship between diabetes and cutaneous candidiasis necessitates a comprehensive understanding to strategize effective management planning. Further investigation and interdisciplinary collaborative efforts are crucial to address this multifaceted challenge and uncover novel approaches for the treatment, management and prevention of both health conditions, including the development of safer and more effective antifungal agents.


Sujet(s)
Antifongiques , Candida albicans , Candidose cutanée , Complications du diabète , Humains , Candida albicans/pathogénicité , Complications du diabète/microbiologie , Candidose cutanée/microbiologie , Candidose cutanée/traitement médicamenteux , Antifongiques/usage thérapeutique , Femelle , Mâle , Diabète/microbiologie , Facteurs de risque , Peau/microbiologie , Peau/anatomopathologie , Prévalence
20.
Sheng Wu Yi Xue Gong Cheng Xue Za Zhi ; 41(3): 635-640, 2024 Jun 25.
Article de Chinois | MEDLINE | ID: mdl-38932552

RÉSUMÉ

Slow wound healing has been a troublesome problem in clinic. In China, traditional methods such as antibiotics and silver sulfadiazine are used to treat skin wound, but the abuse use has many disadvantages, such as chronic wounds and pathogen resistance. Studies have shown that the microorganisms with symbiotic relationship with organisms have benefits on skin wound. Therefore, the way to develop and utilize probiotics to promote wound healing has become a new research direction. In this paper, we reviewed the studies on the bacteriotherapy in the world, described how the probiotics can play a role in promoting wound healing through local wound and intestine, and introduced some mature probiotics products and clinical trials, aiming to provide foundations for further development of bacteriotherapy and products.


Sujet(s)
Probiotiques , Cicatrisation de plaie , Probiotiques/usage thérapeutique , Humains , Peau/microbiologie , Intestins/microbiologie
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...