Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 866
Filter
1.
Acta Derm Venereol ; 104: adv34882, 2024 Jun 11.
Article in English | MEDLINE | ID: mdl-38860624

ABSTRACT

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.


Subject(s)
Dermatitis, Atopic , Skin , Staphylococcus aureus , Superantigens , Humans , Dermatitis, Atopic/microbiology , Superantigens/genetics , Staphylococcus aureus/genetics , Staphylococcus aureus/isolation & purification , Retrospective Studies , Skin/microbiology , Male , Female , Case-Control Studies , Adult , Staphylococcal Skin Infections/microbiology , Middle Aged , Young Adult
2.
Front Immunol ; 15: 1352704, 2024.
Article in English | MEDLINE | ID: mdl-38895118

ABSTRACT

Background: Atopic dermatitis (AD) is a chronic, relapsing inflammatory skin disease with skin barrier defects and a misdirected type 2 immune response against harmless antigens. The skin microbiome in AD is characterized by a reduction in microbial diversity with a dominance of staphylococci, including Staphylococcus epidermidis (S. epidermidis). Objective: To assess whether S. epidermidis antigens play a role in AD, we screened for candidate allergens and studied the T cell and humoral immune response against the extracellular serine protease (Esp). Methods: To identify candidate allergens, we analyzed the binding of human serum IgG4, as a surrogate of IgE, to S. epidermidis extracellular proteins using 2-dimensional immunoblotting and mass spectrometry. We then measured serum IgE and IgG1 binding to recombinant Esp by ELISA in healthy and AD individuals. We also stimulated T cells from AD patients and control subjects with Esp and measured the secreted cytokines. Finally, we analyzed the proteolytic activity of Esp against IL-33 and determined the cleavage sites by mass spectrometry. Results: We identified Esp as the dominant candidate allergen of S. epidermidis. Esp-specific IgE was present in human serum; AD patients had higher concentrations than controls. T cells reacting to Esp were detectable in both AD patients and healthy controls. The T cell response in healthy adults was characterized by IL-17, IL-22, IFN-γ, and IL-10, whereas the AD patients' T cells lacked IL-17 production and released only low amounts of IL-22, IFN-γ, and IL-10. In contrast, Th2 cytokine release was higher in T cells from AD patients than from healthy controls. Mature Esp cleaved and activated the alarmin IL-33. Conclusion: The extracellular serine protease Esp of S. epidermidis can activate IL-33. As an antigen, Esp elicits a type 2-biased antibody and T cell response in AD patients. This suggests that S. epidermidis can aggravate AD through the allergenic properties of Esp.


Subject(s)
Dermatitis, Atopic , Immunoglobulin E , Serine Proteases , Staphylococcus epidermidis , Humans , Staphylococcus epidermidis/immunology , Dermatitis, Atopic/immunology , Dermatitis, Atopic/microbiology , Serine Proteases/immunology , Serine Proteases/metabolism , Adult , Male , Female , Immunoglobulin E/immunology , Immunoglobulin E/blood , Bacterial Proteins/immunology , Immunoglobulin G/immunology , Immunoglobulin G/blood , Cytokines/metabolism , Cytokines/immunology , T-Lymphocytes/immunology , Allergens/immunology , Interleukin-33/immunology , Middle Aged
3.
Exp Dermatol ; 33(6): e15108, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38855891

ABSTRACT

Head and neck atopic dermatitis (HNAD) is a subtype of atopic dermatitis (AD), a common inflammatory skin condition with a distinctive clinical appearance. Malassezia spp., a predominant skin yeast, is considered to exacerbate HNAD. In this study, we investigate the prevalence of Malassezia-specific IgE among HNAD patients. A comprehensive search was performed for observational studies analysing the association between Malassezia-specific IgE and HNAD. This study was performed according to the Preferred Reporting Items for Systematic reviews and Meta-Analyses 2020 checklist and quality was assessed via the Newcastle-Ottawa Quality Assessment Scale (NOS). Fourteen observational studies (840 patients) were included in the analysis. 58% of HNAD patients were male (95% CI: 45.2-69.7). Overall prevalence of Malassezia-specific IgE among HNAD patients was 79.3% (95% CI: 57.5-91.5). Prevalence of Malassezia-specific IgE among HNAD patients varied significantly between geographical regions (p = 0.0441), with 88% in non-Asian regions (95% CI: 61.06-97.17) and 54.73% in Asian regions (95% CI: 34.36-73.63). Malassezia-specific IgE prevalence among HNAD patients varied significantly among studies of higher and lower NOS quality score (p = 0.0386), with 95.42% in studies with NOS ≥7 (95% CI: 63.54-99.60) and 58.05% in studies with NOS <7 (95% CI: 41.44-73.01). Malassezia-specific IgE prevalence among HNAD patients did not vary significantly between more and less predominant Malassezia species (p = 0.1048). Malassezia spp. plays a crucial role in the pathogenesis of HNAD, and IgE anti-Malassezia antibodies appeared to be a common marker for HNAD. Understanding the pathophysiology of Malassezia in HNAD can help develop more targeted therapeutic approaches in managing AD.


Subject(s)
Dermatitis, Atopic , Immunoglobulin E , Malassezia , Malassezia/immunology , Humans , Immunoglobulin E/blood , Dermatitis, Atopic/microbiology , Dermatitis, Atopic/immunology , Prevalence , Eczema/immunology , Eczema/microbiology , Male , Neck/microbiology , Female , Head/microbiology
4.
Skin Res Technol ; 30(7): e13792, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38940462

ABSTRACT

BACKGROUND: Atopic dermatitis (AD) is a chronic inflammatory skin disease that affects 15%-30% of children and 10% of adults globally, with its incidence being influenced by genetic, environmental, and various other factors. While the immune plays a crucial role in the development, the composition of gut microbiota and serum metabolites also contribute to its pathogenesis. SUBJECT: Study the characteristics of gut microbiota and serum metabolites in patients with atopic dermatitis METHOD: In this study, we collected stool and serum samples from 28 AD patients and 23 healthy individuals (NC) for metagenomic sequencing of gut microbiota and non-targeted metabolomic sequencing of serum. RESULT: Our results revealed a lower diversity of gut microbiota in the AD group compared to the NC group. The predominant Phylum in AD patients were Bacteroidetes, Pseudomonas, and Verrucomicrobia, with the most dominant bacterial genus being Faecalibacterium. At the species level, Prevotella copri and Faecalibacterium prausnitzii were found to be the most abundant bacteria. Significant differences in serum metabolite profiles were observed between NC and AD patients, with noticeable variations in metabolite expression levels. The majority of metabolites in the serum of AD patients exhibited low expression, while a few showed high expression levels. Notably, metabolites such as Cholesterol glucuronide, Styrene, Lutein, Betaine, Phosphorylcholine, Taurine, and Creatinine displayed the most pronounced alterations. CONCLUSION: These findings contribute to a further understanding of the complexities underlying this disease.


Subject(s)
Dermatitis, Atopic , Feces , Gastrointestinal Microbiome , Humans , Dermatitis, Atopic/microbiology , Dermatitis, Atopic/blood , Gastrointestinal Microbiome/physiology , Male , Female , Adult , Feces/microbiology , Child , Young Adult , Middle Aged , Adolescent , Metabolome/physiology , Bacteroidetes
5.
Int J Mol Sci ; 25(12)2024 Jun 13.
Article in English | MEDLINE | ID: mdl-38928245

ABSTRACT

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.


Subject(s)
Dermatitis, Atopic , Dysbiosis , Gastrointestinal Microbiome , Probiotics , Dermatitis, Atopic/microbiology , Dermatitis, Atopic/therapy , Humans , Dysbiosis/microbiology , Dysbiosis/therapy , Probiotics/therapeutic use , Microbiota , Skin/microbiology , Animals
6.
Mycoses ; 67(6): e13751, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38825584

ABSTRACT

BACKGROUND: Kerion is a severe type of tinea capitis that is difficult to treat and remains a public health problem. OBJECTIVES: To evaluate the epidemiologic features and efficacy of different treatment schemes from real-world experience. METHODS: From 2019 to 2021, 316 patients diagnosed with kerion at 32 tertiary Chinese hospitals were enrolled. We analysed the data of each patient, including clinical characteristics, causative pathogens, treatments and outcomes. RESULTS: Preschool children were predominantly affected and were more likely to have zoophilic infection. The most common pathogen in China was Microsporum canis. Atopic dermatitis (AD), animal contact, endothrix infection and geophilic pathogens were linked with kerion occurrence. In terms of treatment, itraconazole was the most applied antifungal agent and reduced the time to mycological cure. A total of 22.5% of patients received systemic glucocorticoids simultaneously, which reduced the time to complete symptom relief. Furthermore, glucocorticoids combined with itraconazole had better treatment efficacy, with a higher rate and shorter time to achieving mycological cure. CONCLUSIONS: Kerion often affects preschoolers and leads to serious sequelae, with AD, animal contact, and endothrix infection as potential risk factors. Glucocorticoids, especially those combined with itraconazole, had better treatment efficacy.


Subject(s)
Antifungal Agents , Itraconazole , Microsporum , Tinea Capitis , Humans , Child, Preschool , Antifungal Agents/therapeutic use , Male , Female , Tinea Capitis/drug therapy , Tinea Capitis/epidemiology , Tinea Capitis/microbiology , Itraconazole/therapeutic use , China/epidemiology , Microsporum/isolation & purification , Child , Infant , Glucocorticoids/therapeutic use , Treatment Outcome , Dermatitis, Atopic/drug therapy , Dermatitis, Atopic/epidemiology , Dermatitis, Atopic/microbiology , Risk Factors , Adolescent , Adult , Middle Aged , Retrospective Studies
7.
J Dermatol Sci ; 114(3): 104-114, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38806322

ABSTRACT

BACKGROUND: Although several mouse models of exogenous-agent-induced atopic dermatitis (AD) are currently available, the lack of certainty regarding their similarity with human AD has limited their scientific value. Thus, comprehensive evaluation of the characteristics of mouse models and their similarity with human AD is essential. OBJECTIVE: To compare six different exogenous-agent-induced AD mouse models and find out the optimum models for study. METHODS: Female BALB/c mice underwent induction of AD-like dermatitis by MC903 alone or in combination with ovalbumin (OVA), dinitrofluorobenzene (DNFB) alone or in combination with OVA, OVA alone, or Staphylococcus aureus. Gross phenotype, total immunoglobulin E (IgE) level, histopathological manifestations, and skin lesion transcriptome were analyzed, and metagenomic sequencing of the gut microbiome was performed. RESULTS: The DNFB plus OVA model showed the highest disease severity, while the OVA model showed the lowest severity. The MC903 and MC903 plus OVA models showed high expression of T-helper (Th)2- and Th17-related genes; the DNFB and DNFB plus OVA models showed upregulation of Th1-, Th2-, and Th17-related genes; while the S. aureus inoculation model showed more enhanced Th1 and Th17 immune responses. In contrast to the other models, the OVA-induced model showed the lowest expression levels of inflammation-related genes, while the MC903 model shared the largest overlap with human AD profiles. The intestinal microbiota of all groups showed significant differences after modeling. CONCLUSION: Each AD mouse model exhibited different characteristics. The MC903 model was the best to recapitulate most features of human AD among these exogenous-agent-induced AD models.


Subject(s)
Dermatitis, Atopic , Dinitrofluorobenzene , Disease Models, Animal , Mice, Inbred BALB C , Ovalbumin , Phenotype , Staphylococcus aureus , Transcriptome , Animals , Dermatitis, Atopic/immunology , Dermatitis, Atopic/microbiology , Dermatitis, Atopic/chemically induced , Female , Mice , Ovalbumin/immunology , Staphylococcus aureus/immunology , Humans , Skin/immunology , Skin/pathology , Skin/microbiology , Immunoglobulin E/blood , Immunoglobulin E/immunology , Gastrointestinal Microbiome/immunology , Severity of Illness Index , Gene Expression Profiling , Calcitriol/analogs & derivatives
8.
JCI Insight ; 9(9)2024 Apr 11.
Article in English | MEDLINE | ID: mdl-38716729

ABSTRACT

Atopic dermatitis (AD) is an inflammatory skin condition with a childhood prevalence of up to 25%. Microbial dysbiosis is characteristic of AD, with Staphylococcus aureus the most frequent pathogen associated with disease flares and increasingly implicated in disease pathogenesis. Therapeutics to mitigate the effects of S. aureus have had limited efficacy and S. aureus-associated temporal disease flares are synonymous with AD. An alternative approach is an anti-S. aureus vaccine, tailored to AD. Experimental vaccines have highlighted the importance of T cells in conferring protective anti-S. aureus responses; however, correlates of T cell immunity against S. aureus in AD have not been identified. We identify a systemic and cutaneous immunological signature associated with S. aureus skin infection (ADS.aureus) in a pediatric AD cohort, using a combined Bayesian multinomial analysis. ADS.aureus was most highly associated with elevated cutaneous chemokines IP10 and TARC, which preferentially direct Th1 and Th2 cells to skin. Systemic CD4+ and CD8+ T cells, except for Th2 cells, were suppressed in ADS.aureus, particularly circulating Th1, memory IL-10+ T cells, and skin-homing memory Th17 cells. Systemic γδ T cell expansion in ADS.aureus was also observed. This study suggests that augmentation of protective T cell subsets is a potential therapeutic strategy in the management of S. aureus in AD.


Subject(s)
Dermatitis, Atopic , Staphylococcal Skin Infections , Staphylococcus aureus , Dermatitis, Atopic/immunology , Dermatitis, Atopic/microbiology , Humans , Staphylococcus aureus/immunology , Child , Female , Staphylococcal Skin Infections/immunology , Staphylococcal Skin Infections/microbiology , Male , Child, Preschool , Skin/microbiology , Skin/immunology , Skin/pathology , Chemokine CXCL10/immunology , Chemokine CXCL10/metabolism , Th1 Cells/immunology , Th2 Cells/immunology , Th17 Cells/immunology , Bayes Theorem , CD8-Positive T-Lymphocytes/immunology , Interleukin-10/metabolism , Interleukin-10/immunology , Intraepithelial Lymphocytes/immunology , Antigens, Differentiation, T-Lymphocyte , Membrane Glycoproteins
9.
Adv Exp Med Biol ; 1447: 227-244, 2024.
Article in English | MEDLINE | ID: mdl-38724797

ABSTRACT

This chapter thoroughly examines recent breakthroughs in atopic dermatitis (AD) treatment, with a primary focus on the medications in the development pipeline. Biologics agents targeting new interleukin receptors like interleukin-31, interleukin-22, and interleukin-2 are discussed along with the novel pathway looking at the OX40-OX40L interaction. Oral agents and small molecule therapies like Janus kinase inhibitors, sphingosine-1-phosphate modulators, and Bruton's tyrosine kinase inhibitors are also discussed along with the various new topical medications. Newly approved topicals like phosphodiesterase-4 and JAK inhibitors are highlighted while also discussing the potential of tapinarof and emerging microbiome-targeted therapies. Beyond conventional approaches, the chapter touches upon unconventional therapies currently being studied. The goal of this chapter is to discuss new advances in AD treatment from medications in the initial stages of development to those nearing FDA approval.


Subject(s)
Dermatitis, Atopic , Dermatitis, Atopic/drug therapy , Dermatitis, Atopic/therapy , Dermatitis, Atopic/microbiology , Dermatitis, Atopic/immunology , Humans , Biological Products/therapeutic use , Phosphodiesterase 4 Inhibitors/therapeutic use , Janus Kinase Inhibitors/therapeutic use , Animals , Molecular Targeted Therapy/methods , Dermatologic Agents/therapeutic use
10.
Int J Mol Sci ; 25(10)2024 May 10.
Article in English | MEDLINE | ID: mdl-38791249

ABSTRACT

Atopic dermatitis (AD) is a chronic inflammatory skin disorder influenced by genetic predisposition, environmental factors, immune dysregulation, and skin barrier dysfunction. The skin microbiome and metabolome play crucial roles in modulating the skin's immune environment and integrity. However, their specific contributions to AD remain unclear. We aimed to investigate the distinct skin microbial communities and skin metabolic compounds in AD patients compared to healthy controls (HCs). Seven patients with AD patients and seven HCs were enrolled, from whom skin samples were obtained for examination. The study involved 16S rRNA metagenomic sequencing and bioinformatics analysis as well as the use of gas chromatography time-of-flight mass spectrometry (GC-TOF-MS) to detect metabolites associated with AD in the skin. We observed significant differences in microbial diversity between lesional and non-lesional skin of AD patients and HCs. Staphylococcus overgrowth was prominent in AD lesions, while Cutibacterium levels were decreased. Metabolomic analysis revealed elevated levels of several metabolites, including hypoxanthine and glycerol-3-phosphate in AD lesions, indicating perturbations in purine metabolism and energy production pathways. Moreover, we found a positive correlation between hypoxanthine and glycerol-3-phosphate and clinical severity of AD and Staphylococcus overgrowth. These findings suggest potential biomarkers for monitoring AD severity. Further research is needed to elucidate the causal relationships between microbial dysbiosis, metabolic alterations, and AD progression, paving the way for targeted therapeutic interventions.


Subject(s)
Dermatitis, Atopic , Metabolome , Microbiota , Skin , Dermatitis, Atopic/microbiology , Dermatitis, Atopic/metabolism , Humans , Skin/microbiology , Skin/metabolism , Female , Male , Adult , RNA, Ribosomal, 16S/genetics , Metabolomics/methods , Young Adult , Middle Aged , Case-Control Studies
11.
PLoS One ; 19(5): e0298361, 2024.
Article in English | MEDLINE | ID: mdl-38814946

ABSTRACT

The pathogenesis of anal sacculitis has not been extensively investigated, although atopic dogs seem to be predisposed to the disease. The aim of this study was therefore to characterize and compare the bacterial microbiota and pro-inflammatory cytokines in the anal sacs of dogs from three groups (healthy dogs, untreated atopic dogs and atopic dogs receiving antipruritic treatment or allergen-specific immunotherapy) in order to determine whether changes could be at the origin of anal sacculitis in atopic dogs. Bacterial populations of anal sac secretions from fifteen healthy dogs, fourteen untreated and six treated atopic dogs were characterized by sequencing the V4 region of the 16S rRNA gene using Illumina technology. Proinflammatory cytokines were analyzed with the Luminex multiplex test. Community membership and structure were significantly different between the anal sacs of healthy and untreated atopic dogs (P = 0.002 and P = 0.003, respectively) and between those of untreated and treated atopic dogs (P = 0.012 and P = 0.017, respectively). However, the community structure was similar in healthy and treated atopic dogs (P = 0.332). Among the proinflammatory cytokines assessed, there was no significant difference between groups, except for interleukin 8 which was higher in the anal sacs of untreated atopic dogs compared to treated atopic dogs (P = 0.02), and tumor necrosis factor-alpha which was lower in the anal sacs of healthy dogs compared to treated atopic dogs (P = 0.04). These results reveal a dysbiosis in the anal sacs of atopic dogs, which may partially explain the predisposition of atopic dogs to develop bacterial anal sacculitis. Treatments received by atopic dogs (oclacitinib, desloratadine and allergen-specific immunotherapy) shift the microbiota of the anal sacs towards that of healthy dogs. Further studies are required to identify significant cytokines contributing to anal sacculitis in atopic dogs.


Subject(s)
Anal Sacs , Cytokines , Dog Diseases , Animals , Dogs , Cytokines/metabolism , Dog Diseases/microbiology , Dog Diseases/immunology , Dog Diseases/drug therapy , Anal Sacs/microbiology , Male , Microbiota , Female , RNA, Ribosomal, 16S/genetics , Dermatitis, Atopic/veterinary , Dermatitis, Atopic/microbiology , Dermatitis, Atopic/drug therapy , Dermatitis, Atopic/immunology , Case-Control Studies , Bacteria/classification , Bacteria/genetics
12.
FEMS Microbiol Ecol ; 100(6)2024 May 14.
Article in English | MEDLINE | ID: mdl-38806244

ABSTRACT

Coagulase-negative Staphylococcus (CoNS) species inhibiting Staphylococcus aureus has been described in the skin of atopic dermatitis (AD) patients. This study evaluated whether Staphylococcus spp. from the skin and nares of AD and non-AD children produced antimicrobial substances (AMS). AMS production was screened by an overlay method and tested against NaOH, proteases and 30 indicator strains. Clonality was assessed by pulsed-field gel electrophoresis. Proteinaceous AMS-producers were investigated for autoimmunity by the overlay method and presence of bacteriocin genes by polymerase chain reaction. Two AMS-producers had their genome screened for AMS genes. A methicillin-resistant S. aureus (MRSA) produced proteinaceous AMS that inhibited 51.7% of the staphylococcal indicator strains, and it was active against 60% of the colonies selected from the AD child where it was isolated. On the other hand, 57 (8.8%) CoNS from the nares and skin of AD and non-AD children, most of them S. epidermidis (45.6%), reduced the growth of S. aureus and other CoNS species. Bacteriocin-related genes were detected in the genomes of AMS-producers. AMS production by CoNS inhibited S. aureus and other skin microbiota species from children with AD. Furthermore, an MRSA colonizing a child with AD produced AMS, reinforcing its contribution to dysbiosis and disease severity.


Subject(s)
Coagulase , Dermatitis, Atopic , Methicillin-Resistant Staphylococcus aureus , Microbiota , Skin , Staphylococcus , Dermatitis, Atopic/microbiology , Humans , Methicillin-Resistant Staphylococcus aureus/genetics , Skin/microbiology , Child , Coagulase/genetics , Coagulase/metabolism , Staphylococcus/genetics , Bacteriocins/genetics , Anti-Bacterial Agents/pharmacology , Child, Preschool , Microbial Sensitivity Tests
13.
Adv Exp Med Biol ; 1447: 117-129, 2024.
Article in English | MEDLINE | ID: mdl-38724789

ABSTRACT

With recent advances in topical therapies for atopic dermatitis (AD), steroid-sparing options like calcineurin inhibitors, Janus kinase (JAK) inhibitors, and phosphodiesterase-4 (PDE-4) inhibitors are becoming mainstays in therapy, underscoring the importance of careful selection and usage of topical corticosteroids (TCSs) to minimize side effects. Alongside the necessity of emollient use, these steroid-sparing alternatives offer rapid itch relief and efficacy in improving disease severity. While TCSs still hold a prominent role in AD management, promising novel topical treatments like tapinarof and live biotherapeutics to modulate the skin microbiome are also discussed. Overall, the recent addition of novel topical therapies offers diverse options for AD management and underscores the importance of topical treatments in the management of AD.


Subject(s)
Dermatitis, Atopic , Humans , Dermatitis, Atopic/drug therapy , Dermatitis, Atopic/microbiology , Administration, Topical , Adrenal Cortex Hormones/therapeutic use , Adrenal Cortex Hormones/administration & dosage , Phosphodiesterase 4 Inhibitors/therapeutic use , Administration, Cutaneous , Skin/drug effects , Skin/microbiology , Skin/pathology , Calcineurin Inhibitors/therapeutic use , Calcineurin Inhibitors/administration & dosage , Dermatologic Agents/therapeutic use , Dermatologic Agents/administration & dosage , Dermatologic Agents/adverse effects
14.
Am J Clin Dermatol ; 25(4): 623-637, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38578398

ABSTRACT

Secondary bacterial infections of common dermatoses such as atopic dermatitis, ectoparasitosis, and varicella zoster virus infections are frequent, with Staphylococcus aureus and Streptococcus pyogenes being the bacteria most involved. There are also Gram-negative infections secondary to common dermatoses such as foot dyshidrotic eczema and tinea pedis. Factors favoring secondary bacterial infections in atopic dermatitis, ectoparasitosis, and varicella zoster virus infections mainly include an epidermal barrier alteration as well as itch. Mite-bacteria interaction is also involved in scabies and some environmental factors can promote Gram-negative bacterial infections of the feet. Furthermore, the bacterial ecology of these superinfections may depend on the geographical origin of the patients, especially in ectoparasitosis. Bacterial superinfections can also have different clinical aspects depending on the underlying dermatoses. Subsequently, the choice of class, course, and duration of antibiotic treatment depends on the severity of the infection and the suspected bacteria, primarily targeting S. aureus. Prevention of these secondary bacterial infections depends first and foremost on the management of the underlying skin disorder. At the same time, educating the patient on maintaining good skin hygiene and reporting changes in the primary lesions is crucial. In the case of recurrent secondary infections, decolonization of S. aureus is deemed necessary, particularly in atopic dermatitis.


Subject(s)
Anti-Bacterial Agents , Dermatitis, Atopic , Humans , Dermatitis, Atopic/complications , Dermatitis, Atopic/microbiology , Dermatitis, Atopic/diagnosis , Anti-Bacterial Agents/therapeutic use , Superinfection/microbiology , Superinfection/diagnosis , Skin Diseases, Bacterial/microbiology , Skin Diseases, Bacterial/diagnosis , Skin Diseases, Bacterial/epidemiology , Staphylococcus aureus/isolation & purification , Coinfection/microbiology
15.
Nutrients ; 16(8)2024 Apr 20.
Article in English | MEDLINE | ID: mdl-38674918

ABSTRACT

As gluten may trigger gastrointestinal disorders (GIDs), its presence or absence in the diet can change the diversity and proportion of gut microbiota. The effects of gluten after six weeks of a double-blind, placebo-controlled intervention with a gluten-free diet (GFD) were studied in participants with GIDs suffering from migraines and atopic dermatitis (n = 46). Clinical biomarkers, digestive symptoms, stool, the Migraine Disability Assessment questionnaire, and zonulin levels were analyzed. Next-generation sequencing was used to amplify the 16S rRNA gene of bacteria and the internal transcribed spacer (ITS) regions of fungi. The GFD increased Chao1 fungal diversity after the intervention, while the fungal composition showed no changes. Bacterial diversity and composition remained stable, but a positive association between bacterial and fungal Chao1 diversity and a negative association between Dothideomycetes and Akkermansia were observed. GIDs decreased in both groups and migraines improved in the placebo group. Our findings may aid the development of GID treatment strategies.


Subject(s)
Diet, Gluten-Free , Gastrointestinal Diseases , Gastrointestinal Microbiome , Glutens , Migraine Disorders , Humans , Migraine Disorders/microbiology , Female , Male , Gastrointestinal Diseases/microbiology , Adult , Double-Blind Method , Glutens/adverse effects , Middle Aged , Dermatitis, Atopic/microbiology , Feces/microbiology , Bacteria/classification , Bacteria/genetics , Bacteria/isolation & purification , Fungi , RNA, Ribosomal, 16S , Protein Precursors , Haptoglobins
16.
J Invest Dermatol ; 144(5): 1001-1009, 2024 May.
Article in English | MEDLINE | ID: mdl-38573278

ABSTRACT

Whereas clinically apparent atopic dermatitis (AD) can be confirmed by validated diagnostic criteria, the preclinical phenotype of infants who eventually develop AD is less well-characterized. Analogous to unaffected or nonlesional skin in established AD, clinically normal-appearing skin in infants who will develop clinical AD has distinct changes. Prospective studies have revealed insights into this preclinical AD phenotype. In this study, we review the structural, immunologic, and microbiome nature of the preclinical AD phenotype. Determination of markers that predict the development of AD will facilitate targeting of interventions to prevent the development or reduce the severity of AD in infants.


Subject(s)
Dermatitis, Atopic , Skin , Humans , Infant , Biomarkers/analysis , Dermatitis, Atopic/immunology , Dermatitis, Atopic/microbiology , Dermatitis, Atopic/diagnosis , Microbiota/immunology , Phenotype , Severity of Illness Index , Skin/microbiology , Skin/immunology , Skin/pathology
17.
J Invest Dermatol ; 144(5): 969-977, 2024 May.
Article in English | MEDLINE | ID: mdl-38530677

ABSTRACT

Atopic dermatitis (AD) is a multifactorial, heterogeneous disease characterized by epidermal barrier dysfunction, immune system dysregulation, and skin microbiome alterations. Skin microbiome studies in AD have demonstrated that disease flares are associated with microbial shifts, particularly Staphylococcus aureus predominance. AD-associated S. aureus strains differ from those in healthy individuals across various genomic loci, including virulence factors, adhesion proteins, and proinflammatory molecules-which may contribute to complex microbiome barrier-immune system interactions in AD. Different microbially based treatments for AD have been explored, and their future therapeutic successes will depend on a deeper understanding of the potential microbial contributions to the disease.


Subject(s)
Dermatitis, Atopic , Microbiota , Skin , Staphylococcus aureus , Humans , Dermatitis, Atopic/microbiology , Dermatitis, Atopic/immunology , Microbiota/immunology , Skin/microbiology , Skin/immunology , Staphylococcus aureus/immunology
18.
J Invest Dermatol ; 144(5): 950-953, 2024 May.
Article in English | MEDLINE | ID: mdl-38430083

ABSTRACT

Pruritus or itch is a defining symptom of atopic dermatitis (AD). The origins of itch are complex, and it is considered both a defense mechanism and a cause of disease that leads to inflammation and psychological stress. Considerable progress has been made in understanding the processes that trigger itch, particularly the pruritoceptive origins that are generated in the skin. This perspective review discusses the implications of a recent observation that the V8 protease expressed by Staphylococcus aureus can directly trigger sensory neurons in the skin through activation of protease-activated receptor 1. This may be a key to understanding why itch is so common in AD because S. aureus commonly overgrows in this disease owing to deficient antimicrobial defense from both the epidermis and the cutaneous microbiome. Increased understanding of the role of microbes in AD provides increased opportunities for safely improving the treatment of this disorder.


Subject(s)
Dermatitis, Atopic , Pruritus , Staphylococcus aureus , Dermatitis, Atopic/microbiology , Dermatitis, Atopic/complications , Dermatitis, Atopic/immunology , Humans , Pruritus/microbiology , Pruritus/immunology , Animals , Skin/microbiology , Skin/pathology , Sensory Receptor Cells/physiology , Sensory Receptor Cells/metabolism , Receptors, Proteinase-Activated/metabolism , Staphylococcal Infections/complications , Staphylococcal Infections/microbiology
19.
Sci Rep ; 14(1): 3168, 2024 02 07.
Article in English | MEDLINE | ID: mdl-38326388

ABSTRACT

Altered gut microbiome composition has been reported in children with eczema and interventions that restore beneficial bacteria in the gut may improve eczema. This open-label pilot study aimed to investigate the efficacy of a novel infant microbiome formula (SIM03) in young children with eczema. Pre-school Chinese children aged 1-5 years old with eczema received SIM03 twice daily for three months. The novelty of SIM03 consists of both the use of a patented microencapsulation technology to protect the viability of unique Bifidobacterium bifidum and Bifidobacterium breve strains identified through big data analysis of large metagenomic datasets of young Chinese children. Paired stool samples at baseline and following SIM03 were analyzed by metagenomics sequencing. Generalized estimating equation was used to analyze changes in eczema severity, skin biophysical parameters, quality of life and stool microbiome. Twenty children aged 3.0 ± 1.6 years (10 with severe eczema) were recruited. Treatment compliance was ≥ 98%. SCORing Atopic Dermatitis score decreased significantly at two months (P = 0.008) and three months (P < 0.001), while quality of life improved significantly at 1, 2, and 3 months. The relative abundance of B. breve and microbial pathways on acetate and acetyl-CoA synthesis were enriched in stool samples at one month (P = 0.0014). Children who demonstrated increased B. bifidum after SIM03 showed improvement in sleep loss (P = 0.045). Relative abundance of B. breve correlated inversely with eczema extent (P = 0.023) and intensity (P = 0.019) only among patients with increased B. breve at Month 3. No serious adverse event was observed. In conclusion, SIM03 is well tolerated. This patented microbiome formula improves disease severity and quality of life in young eczematous children by enhancing the delivery of B. bifidum and B. breve in the gut. SIM03 is a potential treatment option for childhood eczema.


Subject(s)
Bifidobacterium bifidum , Dermatitis, Atopic , Eczema , Gastrointestinal Microbiome , Humans , Infant , Child, Preschool , Child , Quality of Life , Pilot Projects , Dermatitis, Atopic/therapy , Dermatitis, Atopic/microbiology , Gastrointestinal Microbiome/genetics , Eczema/therapy
20.
Adv Healthc Mater ; 13(15): e2303272, 2024 06.
Article in English | MEDLINE | ID: mdl-38412280

ABSTRACT

Atopic dermatitis (AD), a prevalent skin condition often complicated by microbial infection, poses a significant challenge in identifying the responsible pathogen for its effective management. However, a reliable, safe tool for pinpointing the source of these infections remains elusive. In this study, a novel on-site pathogen detection that combines chemically functionalized nanotopology with genetic analysis is proposed to capture and analyze pathogens closely associated with severe atopic dermatitis. The chemically functionalized nanotopology features a 3D hierarchical nanopillar array (HNA) with a functional polymer coating, tailored to isolate target pathogens from infected skin. This innovative nanotopology demonstrates superior pathogenic capture efficiency, favorable entrapment patterns, and non-cytotoxicity. An HNA-assembled stick is utilized to directly retrieve bacteria from infected skin samples, followed by extraction-free quantitative loop-mediated isothermal amplification (direct qLAMP) for validation. To mimic human skin conditions, porcine skin is employed to successfully capture Staphylococcus aureus, a common bacterium exacerbating AD cases. The on-site detection method exhibits an impressive detection limit of 103 cells mL-1. The HNA-assembled stick represents a promising tool for on-site detection of bacteria associated with atopic dermatitis. This innovative approach enables to deepen the understanding of AD pathogenesis and open avenues for more effective management strategies for chronic skin conditions.


Subject(s)
Dermatitis, Atopic , Staphylococcus aureus , Dermatitis, Atopic/microbiology , Staphylococcus aureus/isolation & purification , Humans , Animals , Swine , Nucleic Acid Amplification Techniques/methods , Skin/microbiology , Nanostructures/chemistry , Molecular Diagnostic Techniques
SELECTION OF CITATIONS
SEARCH DETAIL
...