What's the 'Skinny' on Microbiome? Interplay of Immune Cells, Microbes, and Skin Barrier in Health and Disease

Blood ◽  
2014 ◽  
Vol 124 (21) ◽  
pp. SCI-46-SCI-46
Author(s):  
Julie Segre ◽  
Heidi Kong ◽  
Fabio Candotti ◽  
Steven M. Holland ◽  
Alexandra F Freeman ◽  
...  

Abstract Culture-based methods have been the primary techniques used to study microbes inhabiting humans; however, many species are not successfully grown in culture. We performed high throughput genomic sequencing surveys to investigate the topographical and temporal complexity of skin microbial communities from 20 skin sites in healthy adults. Significant differences were observed in the bacterial species predominating in particular microenvironments: sebaceous, moist, and dry. Surveying fungal diversity with genomic sequencing, we determined that core body and arm sites were dominated by Malassezia fungi, with species-level classifications revealing greater topographical resolution between sites. Three foot sites, plantar heel, toenail, and toeweb, exhibited tremendous fungal diversity. Concurrent analysis of bacterial and fungal communities demonstrated that skin physiological attributes and topography differentially shape these two microbial communities. While activated and shaped by microbiota, little is known of how the human immune system regulates the human microbiome, and in turn, how this can result in a disease phenotype. We describe the microbial characteristics of the skin of primary immunodeficiency (PID) patients who share a common phenotype of skin eczema yet have different syndromes arising from monogenic mutations leading to loss of distinct lymphocytic populations. We surveyed the skin microbiomes of 41 individuals with Hyper IgE, Wiskott-Aldrich, and Dedicator of Cytokinesis 8 syndromes and compared them against classical atopic dermatitis (AD) patients and healthy controls at skin sites characteristically affected by eczema, a control site, and a site of pathogen carriage (nares). We found that primary immunodeficiency increases the permissiveness of skin microbial colonization not observed in healthy controls or AD patients. We observed decreased site specificity and longitudinal stability in the PID patients as well as unique colonization by environmental microbiota very rare in healthy or AD controls. We identified taxa correlated and anti-correlated with clinical metamarkers in the PID patients; while Staphylococcus aureus, a known pathogen, was most strongly correlated with disease severity, other staphylococci such as S. haemolyticus and S. epidermidis were significantly overrepresented in the PID individuals. These data provide the first illustration of how PID affects microbial prevalence, diversity, and dynamics in relation to skin disease, gaining insight into host-microbiome interactions and how environmental microbes can uniquely colonize PID patients. This comprehensive survey of the skin microbiome also provided the foundation for analyzing changes in the microbial community associated with common forms of AD, which affect ~15% of U.S. children and ~2% of adults and is associated with Staphylococcus aureus colonization and infection. We studied 10 children with moderate to severe AD at baseline, flare, and post-flare, and healthy controls. Severity was quantified using scoring atopic dermatitis (SCORAD). Samples were obtained from characteristically affected areas, a control site, and nares. Bacterial diversity was dramatically reduced during flare as compared to post-flare and controls. Our studies provide comprehensive characterization of skin microbes in AD and controls confirm the frequent culture-based isolation of S. aureus in AD flares and represent one of the earliest longitudinal investigations of the skin microbiome in a dermatologic disorder. Disclosures No relevant conflicts of interest to declare.

Processes ◽  
2021 ◽  
Vol 9 (5) ◽  
pp. 784
Author(s):  
Chao Wang ◽  
Lin Sun ◽  
Haiwen Xu ◽  
Na Na ◽  
Guomei Yin ◽  
...  

Whole-plant corn silages on family farms were sampled in Erdos (S1), Baotou (S2), Ulanqab (S3), and Hohhot (S4) in North China, after 300 d of ensiling. The microbial communities, metabolites, and aerobic stability were assessed. Lactobacillusbuchneri, Acinetobacter johnsonii, and unclassified Novosphingobium were present at greater abundances than others in S2 with greater bacterial diversity and metabolites. Lactobacillus buchneri, Lactobacillus parafarraginis, Lactobacillus kefiri, and unclassified Lactobacillus accounted for 84.5%, and 88.2%, and 98.3% of bacteria in S1, S3, and S4, respectively. The aerobic stability and fungal diversity were greater in S1 and S4 with greater abundances of unclassified Kazachstania, Kazachstania bulderi, Candida xylopsoci, unclassified Cladosporium, Rhizopus microspores, and Candida glabrata than other fungi. The abundances of unclassified Kazachstania in S2 and K. bulderi in S3 were 96.2% and 93.6%, respectively. The main bacterial species in S2 were L. buchneri, A. johnsonii, and unclassified Novosphingobium; Lactobacillus sp. dominated bacterial communities in S1, S3, and S4. The main fungal species in S1 and S4 were unclassified Kazachstania, K. bulderi, C. xylopsoci, unclassified Cladosporium, R. microspores, and C. glabrata; Kazachstania sp. dominated fungal communities in S2 and S3. The high bacterial diversity aided the accumulation of metabolites, and the broad fungal diversity improved the aerobic stability.


Toxins ◽  
2019 ◽  
Vol 11 (6) ◽  
pp. 311 ◽  
Author(s):  
Supitchaya Traisaeng ◽  
Deron Raymond Herr ◽  
Hsin-Jou Kao ◽  
Tsung-Hsien Chuang ◽  
Chun-Ming Huang

The microbiome is a rich source of metabolites for the development of novel drugs. Butyric acid, for example, is a short-chain fatty acid fermentation metabolite of the skin probiotic bacterium Staphylococcus epidermidis (S. epidermidis). Glycerol fermentation of S. epidermidis resulted in the production of butyric acid and effectively hindered the growth of a Staphylococcus aureus (S. aureus) strain isolated from skin lesions of patients with atopic dermatitis (AD) in vitro and in vivo. This approach, however, is unlikely to be therapeutically useful since butyric acid is malodorous and requires a high concentration in the mM range for growth suppression of AD S. aureus. A derivative of butyric acid, BA–NH–NH–BA, was synthesized by conjugation of two butyric acids to both ends of an –NH–O–NH– linker. BA–NH–NH–BA significantly lowered the concentration of butyric acid required to inhibit the growth of AD S. aureus. Like butyric acid, BA–NH–NH–BA functioned as a histone deacetylase (HDAC) inhibitor by inducing the acetylation of Histone H3 lysine 9 (AcH3K9) in human keratinocytes. Furthermore, BA–NH–NH–BA ameliorated AD S. aureus-induced production of pro-inflammatory interleukin (IL)-6 and remarkably reduced the colonization of AD S. aureus in mouse skin. These results describe a novel derivative of a skin microbiome fermentation metabolite that exhibits anti-inflammatory and S. aureus bactericidal activity.


Author(s):  
Hitham Abduarhman Alghamdi ◽  
Ahmed Behieldin ◽  
Sherif Edris

Abstract Atopic dermatitis (AD) is the main focus of this review which mostly starts with children in early life. Beside the etiological factors like environmental, dietary or medical exposures, Gut-skin axis microbiome studies have an impact to investigate and to understand the relation between the gut microbiome and changes to the skin microbiom and some skin diseases resulting like AD. Infants start forming their microbiome in early life and some researches suggest that this time have a crucial role in development of AD. Balanced bacterial composition is important to maintain healthy skin as the gut microbiome dysbiosis may result in dramatic shifting in the skin microbiome that gives better chance for some bacteria such as Staphylococcus aureus to over prevail which has been reported to contribute in AD development. Among several factors, immunological activity has a strong relation to microbiome changed composition and development of AD. Continuous....  


Author(s):  
Olga B. Tamrazova ◽  
Evgenia A. Glukhova ◽  
Anait V. Tamrazova ◽  
Natalia F. Dubovets

Fast traslate Icon translate Fast traslate Icon translate The progressive increase in the incidence of atopic dermatitis among children, an increase in persistence in adulthood, combined with an inevitable decrease in the quality of life of patients, determine the relevance of studying the mechanisms of the development of this disease not only for dermatology, but also for the entire health care system. Thus, the prerequisites for the emergence of new concepts of pathogenesis and the search for the most effective therapeutic modalities arise. At the moment, atopic dermatitis is considered as the interaction of endogenous (impaired immune response, insufficient function of the epidermal barrier) and exogenous (exposure to allergens, chemical or physical irritants, microorganisms) factors. Environmental factors such as temperature and humidity, genetic makeup, antibiotic use, and good hygiene play a critical role in the maintenance and stability of the skin microbiome. Normally, the skin microbiota is mainly formed by bacteria of the genus Staphylococcus, Propionibacterium, Corynebacterium and Streptococcus. While in patients with AD, in 70% of cases, there is colonization of Staphylococcus aureus on the affected skin, in 39% on intact skin, which secondarily contributes to the development of immune imbalance and increased skin xerosis. This fact determines the importance of the use of basic therapy, which, on the one hand, helps to strengthen the epidermal barrier, and on the other hand, normalizes the microbiome of the skin, reducing the colonization of Staphylococcus aureus.


2019 ◽  
Vol 7 (9) ◽  
pp. 301 ◽  
Author(s):  
Enea Gino Di Domenico ◽  
Ilaria Cavallo ◽  
Bruno Capitanio ◽  
Fiorentina Ascenzioni ◽  
Fulvia Pimpinelli ◽  
...  

Biofilm is the dominant mode of growth of the skin microbiota, which promotes adhesion and persistence in the cutaneous microenvironment, thus contributing to the epidermal barrier function and local immune modulation. In turn, the local immune microenvironment plays a part in shaping the skin microbiota composition. Atopic dermatitis (AD) is an immune disorder characterized by a marked dysbiosis, with a sharp decline of microbial diversity. During AD flares biofilm-growing Staphylococcus aureus emerges as the major colonizer in the skin lesions, in strict association with disease severity. The chronic production of inflammatory cytokines in the skin of AD individuals concurs at supporting S. aureus biofilm overgrowth at the expense of other microbial commensals, subverting the composition of the healthy skin microbiome. The close relationship between the host and microbial biofilm resident in the skin has profound implications on human health, making skin microbiota an attractive target for the therapeutic management of different skin disorders.


2019 ◽  
Vol 11 (490) ◽  
pp. eaat8329 ◽  
Author(s):  
Michael R. Williams ◽  
Stephen K. Costa ◽  
Livia S. Zaramela ◽  
Shadi Khalil ◽  
Daniel A. Todd ◽  
...  

Colonization of the skin by Staphylococcus aureus is associated with exacerbation of atopic dermatitis (AD), but any direct mechanism through which dysbiosis of the skin microbiome may influence the development of AD is unknown. Here, we show that proteases and phenol-soluble modulin α (PSMα) secreted by S. aureus lead to endogenous epidermal proteolysis and skin barrier damage that promoted inflammation in mice. We further show that clinical isolates of different coagulase-negative staphylococci (CoNS) species residing on normal skin produced autoinducing peptides that inhibited the S. aureus agr system, in turn decreasing PSMα expression. These autoinducing peptides from skin microbiome CoNS species potently suppressed PSMα expression in S. aureus isolates from subjects with AD without inhibiting S. aureus growth. Metagenomic analysis of the AD skin microbiome revealed that the increase in the relative abundance of S. aureus in patients with active AD correlated with a lower CoNS autoinducing peptides to S. aureus ratio, thus overcoming the peptides’ capacity to inhibit the S. aureus agr system. Characterization of a S. hominis clinical isolate identified an autoinducing peptide (SYNVCGGYF) as a highly potent inhibitor of S. aureus agr activity, capable of preventing S. aureus–mediated epithelial damage and inflammation on murine skin. Together, these findings show how members of the normal human skin microbiome can contribute to epithelial barrier homeostasis by using quorum sensing to inhibit S. aureus toxin production.


Dermatology ◽  
2020 ◽  
pp. 1-7
Author(s):  
Leszek Blicharz ◽  
Maryla Michalak ◽  
Ksenia Szymanek-Majchrzak ◽  
Grażyna Młynarczyk ◽  
Krzysztof Skowroński ◽  
...  

<b><i>Background:</i></b> Atopic dermatitis is a chronic inflammatory dermatosis with complex pathogenesis. The skin microbiome in atopic dermatitis is dominated by <i>Staphylococcus aureus</i> which shows the ability to produce biofilm. <b><i>Objectives:</i></b> The aim of this work was to assess the influence of <i>S. aureus</i> biofilm on the course of atopic dermatitis. <b><i>Methods:</i></b> Disease severity was evaluated based on the SCORAD index in 56 adult patients with atopic dermatitis. Microtiter plate assay of the propensity to form biofilm was performed on <i>S. aureus</i> strains isolated from the anterior nares, lesional skin, and nonlesional skin. Microbiological results were correlated to the clinical parameters and total IgE concentration. <b><i>Results:</i></b> Biofilm-producing strains of <i>S. aureus</i> were identified in 76.3% (29/38) and 79.1% (34/43) of samples from the anterior nares and lesional skin, respectively (<i>p</i> &#x3e; 0.05), and in 48.5% (16/33) of samples from nonlesional skin (<i>p</i> &#x3c; 0.03). Patients colonized by biofilm-producing strains of <i>S. aureus</i> within the anterior nares showed statistically higher mean values of total and objective SCORAD and its components (extent, dryness), and of the largest extent of skin lesions during the flares in the last year when compared to patients colonized by non-biofilm-producing strains. Carriage of biofilm-producing <i>S. aureus</i> on lesional skin was associated with higher mean values of the extent of skin lesions during stable periods of the disease. <b><i>Conclusions:</i></b> The results of this study may suggest a relationship between the production of biofilm by <i>S. aureus</i> strains colonizing the anterior nares and the course of atopic dermatitis. Biofilm seems crucial for dispersal and persistent colonization of large areas of the skin by this pathogen. Destruction of <i>S. aureus</i> biofilm could positively affect the course of atopic dermatitis.


2019 ◽  
Vol 23 (5) ◽  
pp. 247-260 ◽  
Author(s):  
Mohammed Ramadan ◽  
Samar Solyman ◽  
Mamdouh Yones ◽  
Yasser Abdallah ◽  
Hamada Halaby ◽  
...  

Author(s):  
Veda D. Khadka ◽  
Felix M. Key ◽  
Carolina Romo-González ◽  
Adrián Martínez-Gayosso ◽  
Blanca L. Campos-Cabrera ◽  
...  

BackgroundAtopic dermatitis (AD) is characterized by an altered skin microbiome dominantly colonized by S. aureus. Standard treatment includes emollients, anti-inflammatory medications and antiseptics.ObjectivesTo characterize changes in the skin microbiome during treatment for AD.MethodsThe skin microbiomes of children with moderate-to-severe AD and healthy children were investigated in a longitudinal prospective study. Patients with AD were randomized to receive either standard treatment with emollients and topical corticosteroids or standard treatment with the addition of dilute bleach baths (DBB) and sampled at four visits over a three-month period. At each visit, severity of AD was measured, swabs were taken from four body sites and the composition of the microbiome at those sites was assessed using 16S rRNA amplification.ResultsWe included 14 healthy controls and 28 patients. We found high relative abundances of S. aureus in patients, which correlated with AD severity and reduced apparent alpha diversity. As disease severity improved with treatment, the abundance of S. aureus decreased, gradually becoming more similar to the microbiomes of healthy controls. After treatment, patients who received DBB had a significantly lower abundance of S. aureus than those who received only standard treatment.ConclusionsThere are clear differences in the skin microbiome of healthy controls and AD patients that diminish with treatment. After three months, the addition of DBB to standard treatment had significantly decreased the S. aureus burden, supporting its use as a therapeutic option. Further study in double-blinded trials is needed.


Sign in / Sign up

Export Citation Format

Share Document