Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 291
Filter
1.
BMC Microbiol ; 23(1): 273, 2023 09 29.
Article in English | MEDLINE | ID: mdl-37773096

ABSTRACT

There has been considerable research into the understanding of the healthy skin microbiome. Similarly, there is also a considerable body of research into whether specific microbes contribute to skin disorders, with atopic dermatitis (AD) routinely linked to increased Staphylococcus aureus (S. aureus) colonisation. In this study, the epidermal surface of participants was sampled using swabs, while serial tape-stripping (35 tapes) was performed to sample through the stratum corneum. Samples were taken from AD patients and healthy controls, and the bacterial communities were profiled by metabarcoding the universal V3-V4 16S rRNA region. Results show that the majority of bacterial richness is located within the outermost layers of the stratum corneum, however there were many taxa that were found almost exclusively at the very outermost layer of the epidermis. We therefore hypothesise that tape-stripping can be performed to investigate the 'core microbiome' of participants by removing environmental contaminants. Interestingly, significant community variation between AD patients and healthy controls was only observable at the epidermal surface, yet a number of individual taxa were found to consistently differ with AD status across the entire epidermis (i.e. both the epidermal surface and within the epidermis). Sampling strategy could therefore be tailored dependent on the hypothesis, with sampling for forensic applications best performed using surface swabs and outer tapes, while profiling sub-surface communities may better reflect host genome and immunological status.


Subject(s)
Dermatitis, Atopic , Humans , Dermatitis, Atopic/microbiology , Staphylococcus aureus/genetics , RNA, Ribosomal, 16S/genetics , Epidermis/microbiology , Skin/microbiology
2.
Cell Death Dis ; 14(1): 15, 2023 01 11.
Article in English | MEDLINE | ID: mdl-36631452

ABSTRACT

Skin infections caused by drug-resistant Staphylococcus aureus occur at high rates nationwide. Mouse primary epidermal organoids (mPEOs) possess stratified histological and morphological characteristics of epidermis and are highly similar to their derived tissue at the transcriptomic and proteomic levels. Herein, the susceptibility of mPEOs to methicillin-resistant S. aureus USA300 infection was investigated. The results show that mPEOs support USA300 colonization and invasion, exhibiting swollen epithelial squamous cells with nuclear necrosis and secreting inflammatory factors such as IL-1ß. Meanwhile mPEOs beneficial to observe the process of USA300 colonization with increasing infection time, and USA300 induces mPEOs to undergo pyroptosis and autophagy. In addition, we performed a drug screen for the mPEO infection model and showed that vancomycin restores cell viability and inhibits bacterial internalization in a concentration-dependent manner. In conclusion, we establish an in vitro skin infection model that contributes to the examination of drug screening strategies and antimicrobial drug mechanisms.


Subject(s)
Methicillin-Resistant Staphylococcus aureus , Organoids , Staphylococcal Infections , Animals , Mice , Drug Evaluation, Preclinical/methods , Epidermis/metabolism , Epidermis/microbiology , Epidermis/pathology , Proteomics , Staphylococcal Infections/drug therapy , Staphylococcal Infections/microbiology , Organoids/metabolism , Organoids/microbiology
3.
Proc Natl Acad Sci U S A ; 119(30): e2203556119, 2022 07 26.
Article in English | MEDLINE | ID: mdl-35867832

ABSTRACT

Skin microbiome sampling is currently performed with tools such as swabs and tape strips to collect microbes from the skin surface. However, these conventional approaches may be unable to detect microbes deeper in the epidermis or in epidermal invaginations. We describe a sampling tool with a depth component, a transepidermal microprojection array (MPA), which captures microbial biomass from both the epidermal surface and deeper skin layers. We leveraged the rapid customizability of 3D printing to enable systematic optimization of MPA for human skin sampling. Evaluation of sampling efficacy on human scalp revealed the optimized MPA was comparable in sensitivity to swab and superior to tape strip, especially for nonstandard skin surfaces. We observed differences in species diversity, with the MPA detecting clinically relevant fungi more often than other approaches. This work delivers a tool in the complex field of skin microbiome sampling to potentially address gaps in our understanding of its role in health and disease.


Subject(s)
Epidermis , Microbiota , Printing, Three-Dimensional , Specimen Handling , Tissue Array Analysis , Epidermis/microbiology , Humans , Specimen Handling/methods
4.
Microbiol Spectr ; 10(3): e0274421, 2022 06 29.
Article in English | MEDLINE | ID: mdl-35532353

ABSTRACT

Atopic dermatitis (AD) is associated with a deficiency of skin lipids, increased populations of Staphylococcus aureus in the microbiome, and structural defects in the stratum corneum (SC), the outermost layer of human skin. However, the pathogenesis of AD is ambiguous, as it is unclear whether observed changes are the result of AD or contribute to the pathogenesis of the disease. Previous studies have shown that S. aureus is capable of permeating across isolated human SC tissue when lipids are depleted to levels consistent with AD conditions. In this study, we expand upon this discovery to determine the mechanisms and implications of bacterial penetration into the SC barrier. Specifically, we establish if bacteria are permeating intercellularly or employing a combination of both inter- and intracellular travel. The mechanical implications of bacterial invasion, lipid depletion, and media immersion are also evaluated using a newly developed, physiologically relevant, temperature-controlled drip chamber. Results reveal for the first time that S. aureus can be internalized by corneocytes, indicating transcellular movement through the tissue during permeation, consistent with previous theoretical models. S. aureus also degrades the mechanical integrity of human SC, particularly when the tissue is partially depleted of lipids. These observed mechanical changes are likely the cause of broken or ruptured tissue seen as exudative lesions in AD flares. This work further highlights the necessity of lipids in skin microbial barrier function. IMPORTANCE Millions of people suffer from the chronic inflammatory skin disease atopic dermatitis (AD), whose symptoms are associated with a deficiency of skin lipids that exhibit antimicrobial functions and increased populations of the opportunistic pathogen Staphylococcus aureus. However, the pathogenesis of AD is ambiguous, and it remains unclear if these observed changes are merely the result of AD or contribute to the pathogenesis of the disease. In this article, we demonstrate the necessity of skin lipids in preventing S. aureus from penetrating the outermost barrier of human skin, thereby causing a degradation in tissue integrity. This bacterial permeation into the viable epidermis could act as an inflammatory trigger of the disease. When coupled with delipidated AD tissue conditions, bacterial permeation can also explain increased tissue fragility, potentially causing lesion formation in AD patients that results in further enhancing bacterial permeability across the stratum corneum and the development of chronic conditions.


Subject(s)
Dermatitis, Atopic , Staphylococcal Infections , Dermatitis, Atopic/metabolism , Dermatitis, Atopic/microbiology , Epidermis/chemistry , Epidermis/metabolism , Epidermis/microbiology , Humans , Lipids/analysis , Skin/microbiology , Staphylococcus aureus/physiology
5.
Sci Rep ; 12(1): 3078, 2022 02 23.
Article in English | MEDLINE | ID: mdl-35197501

ABSTRACT

Dermatophytic pathogens are a source of disturbance to the host microbiome, but the temporal progression of these disturbances is unclear. Here, we determined how Snake Fungal Disease, caused by Ophidiomyces ophidiicola, resulted in disturbance to the host microbiome. To assess disease effects on the microbiome, 22 Common Watersnakes (Nerodia sipedon) were collected and half were inoculated with O. ophidiicola. Epidermal swabs were collected weekly for use in microbiome and pathogen load characterization. For the inoculated treatment only, we found a significant effect of disease progression on microbial richness and Shannon diversity consistent with the intermediate disturbance hypothesis. When explicitly accounting for differences in assemblage richness, we found that ß-diversity among snakes was significantly affected by the interaction of time and treatment group, with assemblages becoming more dissimilar across time in the inoculated, but not the control group. Also, differences between treatments in average microbiome composition became greater with time, but this interactive effect was not evident when accounting for assemblage richness. These results suggest that changes in composition of the host microbiome associated with disease largely occur due to changes in microbial richness related to disease progression.


Subject(s)
Animal Diseases/microbiology , Colubridae/microbiology , Epidermis/microbiology , Host Microbial Interactions/physiology , Mycoses/microbiology , Onygenales/pathogenicity , Animals , Disease Progression , Time Factors
6.
Front Immunol ; 12: 774018, 2021.
Article in English | MEDLINE | ID: mdl-34925344

ABSTRACT

The epidermis constitutes a continuous external layer covering the body, offering protection against bacteria, the most abundant living organisms that come into contact with this barrier. The epidermis is heavily colonized by commensal bacterial organisms that help protect against pathogenic bacteria. The highly regulated and dynamic interaction between the epidermis and commensals involves the host's production of nutritional factors promoting bacterial growth together to chemical and immunological bacterial inhibitors. Signal trafficking ensures the system's homeostasis; conditions that favor colonization by pathogens frequently foster commensal growth, thereby increasing the bacterial population size and inducing the skin's antibacterial response, eliminating the pathogens and re-establishing the normal density of commensals. The microecological conditions of the epidermis favors Gram-positive organisms and are unsuitable for long-term Gram-negative colonization. However, the epidermis acts as the most important host-to-host transmission platform for bacteria, including those that colonize human mucous membranes. Bacteria are frequently shared by relatives, partners, and coworkers. The epidermal bacterial transmission platform of healthcare workers and visitors can contaminate hospitalized patients, eventually contributing to cross-infections. Epidermal transmission occurs mostly via the hands and particularly through fingers. The three-dimensional physical structure of the epidermis, particularly the fingertips, which have frictional ridges, multiplies the possibilities for bacterial adhesion and release. Research into the biology of bacterial transmission via the hands is still in its infancy; however, tribology, the science of interacting surfaces in relative motion, including friction, wear and lubrication, will certainly be an important part of it. Experiments on finger-to-finger transmission of microorganisms have shown significant interindividual differences in the ability to transmit microorganisms, presumably due to genetics, age, sex, and the gland density, which determines the physical, chemical, adhesive, nutritional, and immunological status of the epidermal surface. These studies are needed to optimize interventions and strategies for preventing the hand transmission of microorganisms.


Subject(s)
Bacterial Infections/transmission , Epidermis/microbiology , Bacteria/growth & development , Epidermis/immunology , Fingers/microbiology , Hand/microbiology , Humans , Microbiota
7.
Emerg Microbes Infect ; 10(1): 2326-2339, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34821529

ABSTRACT

ABSTRACTMelioidosis is a serious infectious disease endemic in Southeast Asia, Northern Australia and has been increasingly reported in other tropical and subtropical regions in the world. Percutaneous inoculation through cuts and wounds on the skin is one of the major modes of natural transmission. Despite cuts in skin being a major route of entry, very little is known about how the causative bacterium Burkholderia pseudomallei initiates an infection at the skin and the disease manifestation at the skin known as cutaneous melioidosis. One key issue is the lack of suitable and relevant infection models. Employing an in vitro 2D keratinocyte cell culture, a 3D skin equivalent fibroblast-keratinocyte co-culture and ex vivo organ culture from human skin, we developed infection models utilizing surrogate model organism Burkholderia thailandensis to investigate Burkholderia-skin interactions. Collectively, these models show that the bacterial infection was largely limited at the wound's edge. Infection impedes wound closure, triggers inflammasome activation and cellular extrusion in the keratinocytes as a potential way to control bacterial infectious load at the skin. However, extensive infection over time could result in the epidermal layer being sloughed off, potentially contributing to formation of skin lesions.


Subject(s)
Burkholderia pseudomallei/physiology , Burkholderia/physiology , Epidermis/microbiology , Inflammasomes/metabolism , Keratinocytes/microbiology , Melioidosis/microbiology , Skin/microbiology , Wounds and Injuries/microbiology , Cells, Cultured , Epidermis/metabolism , Humans , Keratinocytes/metabolism , Melioidosis/metabolism , Melioidosis/pathology , Models, Biological , Skin/metabolism , Skin/pathology , Wounds and Injuries/metabolism , Wounds and Injuries/pathology
8.
Int J Mol Sci ; 22(21)2021 Oct 28.
Article in English | MEDLINE | ID: mdl-34769105

ABSTRACT

The epidermis is a living, multilayered barrier with five functional levels, including a physical, a chemical, a microbial, a neuronal, and an immune level. Altogether, this complex organ contributes to protect the host from external aggression and to preserve its integrity. In this review, we focused on the different functional aspects.


Subject(s)
Epidermis/physiology , Epidermis/microbiology , Humans , Immunity , Microbiota , Sensory Receptor Cells/physiology
9.
J Clin Invest ; 131(24)2021 12 15.
Article in English | MEDLINE | ID: mdl-34730110

ABSTRACT

Impaired wound healing associated with recurrent Staphylococcus aureus infection and unresolved inflammation are hallmarks of nonhealing diabetic foot ulcers (DFUs). Perforin-2, an innate immunity molecule against intracellular bacteria, limits cutaneous infection and dissemination of S. aureus in mice. Here, we report the intracellular accumulation of S. aureus in the epidermis of DFUs with no clinical signs of infection due to marked suppression of perforin-2. S. aureus residing within the epidermis of DFUs triggers AIM2 inflammasome activation and pyroptosis. These findings were corroborated in mice lacking perforin-2. The effects of pyroptosis on DFU clinical outcomes were further elucidated in a 4-week longitudinal clinical study in patients with DFUs receiving standard care. Increased AIM2 inflammasome and ASC-pyroptosome coupled with induction of IL-1ß were found in nonhealing DFUs compared with healing DFUs. Our findings revealed that perforin-2 suppression, intracellular S. aureus accumulation, and associated induction of pyroptosis contribute to healing inhibition and prolonged inflammation in patients with DFUs.


Subject(s)
Diabetic Foot/immunology , Epidermis/immunology , Membrane Proteins/immunology , Pore Forming Cytotoxic Proteins/immunology , Pyroptosis/immunology , Staphylococcal Infections/immunology , Staphylococcus aureus/immunology , Wound Healing/immunology , Adult , Aged , Animals , DNA-Binding Proteins/genetics , DNA-Binding Proteins/immunology , Diabetic Foot/genetics , Diabetic Foot/microbiology , Epidermis/microbiology , Female , Humans , Interleukin-1beta/genetics , Interleukin-1beta/immunology , Male , Membrane Proteins/genetics , Mice , Mice, Knockout , Middle Aged , Pore Forming Cytotoxic Proteins/genetics , Pyroptosis/genetics , Staphylococcal Infections/genetics , Wound Healing/genetics
10.
mBio ; 12(5): e0198921, 2021 10 26.
Article in English | MEDLINE | ID: mdl-34517759

ABSTRACT

The stratum corneum is the outermost layer of the epidermis and is thus directly exposed to the environment. It consists mainly of corneocytes, which are keratinocytes in the last stage of differentiation, having neither nuclei nor organelles. However, they retain keratin filaments embedded in filaggrin matrix and possess a lipid envelope which protects the body from desiccation. Despite the desiccated, nutrient-poor, and acidic nature of the skin making it a hostile environment for most microorganisms, this organ is colonized by commensal microbes. Among the classic skin commensals are Propionibacterium acnes and coagulase-negative staphylococci (CoNS) with Staphylococcus epidermidis as a leading species. An as-yet-unanswered question is what enables S. epidermis to colonize skin so successfully. In their recent article, P. D. Fey and his colleagues (P. Roy, A. R. Horswill, and P. D. Fey, mBio 12:e02908-20, 2021, https://doi.org/10.1128/mBio.02908-20) have brought us one step closer to answering this question.


Subject(s)
Bacterial Proteins/metabolism , Membrane Proteins/metabolism , Staphylococcus epidermidis/metabolism , Bacterial Proteins/genetics , Epidermis/microbiology , Humans , Membrane Proteins/genetics , Skin/microbiology , Staphylococcal Infections/microbiology , Staphylococcus epidermidis/genetics , Staphylococcus epidermidis/growth & development
11.
Front Immunol ; 12: 695373, 2021.
Article in English | MEDLINE | ID: mdl-34512625

ABSTRACT

Langerhans cells (LCs) reside in the epidermis where they are poised to mount an antimicrobial response against microbial pathogens invading from the outside environment. To elucidate potential pathways by which LCs contribute to host defense, we mined published LC transcriptomes deposited in GEO and the scientific literature for genes that participate in antimicrobial responses. Overall, we identified 31 genes in LCs that encode proteins that contribute to antimicrobial activity, ten of which were cross-validated in at least two separate experiments. Seven of these ten antimicrobial genes encode chemokines, CCL1, CCL17, CCL19, CCL2, CCL22, CXCL14 and CXCL2, which mediate both antimicrobial and inflammatory responses. Of these, CCL22 was detected in seven of nine transcriptomes and by PCR in cultured LCs. Overall, the antimicrobial genes identified in LCs encode proteins with broad antibacterial activity, including against Staphylococcus aureus, which is the leading cause of skin infections. Thus, this study illustrates that LCs, consistent with their anatomical location, are programmed to mount an antimicrobial response against invading pathogens in skin.


Subject(s)
Antimicrobial Peptides/genetics , Epidermis/metabolism , Langerhans Cells/metabolism , Staphylococcal Skin Infections/genetics , Staphylococcus aureus/pathogenicity , Transcriptome , Cells, Cultured , Databases, Genetic , Epidermis/immunology , Epidermis/microbiology , Gene Expression Profiling , Host-Pathogen Interactions , Humans , Langerhans Cells/immunology , Langerhans Cells/microbiology , Staphylococcal Skin Infections/immunology , Staphylococcal Skin Infections/metabolism , Staphylococcal Skin Infections/microbiology , Staphylococcus aureus/immunology
12.
Virulence ; 12(1): 2474-2492, 2021 12.
Article in English | MEDLINE | ID: mdl-34516337

ABSTRACT

Staphylococcus aureus is a skin commensal microorganism commonly colonizing healthy humans. Nevertheless, S. aureus can also be responsible for cutaneous infections and contribute to flare-up of inflammatory skin diseases such as atopic dermatitis (AD), which is characterized by dysbiosis of the skin microbiota with S. aureus as the predominant species. However, the role of major virulence factors of this pathogen such as phenol-soluble modulin (PSM) toxins in epidermal inflammation remains poorly understood. Stimulation of primary human keratinocytes with sublytic concentrations of synthetic and purified PSM α3 resulted in upregulation of a large panel of pro-inflammatory chemokine and cytokine gene expression, including CXCL1, CXCL2, CXCL3, CXCL5, CXCL8, CCL20, IL-1α, IL-1ß, IL-6, IL-36γ and TNF-α, while inducing the release of CXCL8, CCL20, TNF-α and IL-6. In addition, using S. aureus culture supernatant from mutants deleted from genes encoding either α-type PSMs or all PSM production, PSMs were shown to be the main factors of S. aureus secretome responsible for pro-inflammatory mediator induction in human keratinocytes. On the other hand, α-type PSM-containing supernatant triggered an intense induction of pro-inflammatory mediator expression and secretion during both topical and basal layer stimulation of an ex vivo model of human skin explants, a physiologically relevant model of pluristratified epidermis. Taken together, the results of this study show that PSMs and more specifically α-type PSMs are major virulence factors of S. aureus inducing a potent inflammatory response during infection of the human epidermis and could thereby contribute to AD flare-up through exacerbation of skin inflammation.


Subject(s)
Bacterial Toxins/metabolism , Epidermis , Secretome , Staphylococcal Infections , Staphylococcus aureus , Virulence Factors , Chemokines/immunology , Cytokines/immunology , Epidermis/immunology , Epidermis/microbiology , Humans , Inflammation , Inflammation Mediators/immunology , Staphylococcus aureus/metabolism , Virulence Factors/metabolism
13.
PLoS Genet ; 17(6): e1009600, 2021 06.
Article in English | MEDLINE | ID: mdl-34166401

ABSTRACT

Animals and plants need to defend themselves from pathogen attack. Their defences drive innovation in virulence mechanisms, leading to never-ending cycles of co-evolution in both hosts and pathogens. A full understanding of host immunity therefore requires examination of pathogen virulence strategies. Here, we take advantage of the well-studied innate immune system of Caenorhabditis elegans to dissect the action of two virulence factors from its natural fungal pathogen Drechmeria coniospora. We show that these two enterotoxins have strikingly different effects when expressed individually in the nematode epidermis. One is able to interfere with diverse aspects of host cell biology, altering vesicle trafficking and preventing the key STAT-like transcription factor STA-2 from activating defensive antimicrobial peptide gene expression. The second increases STA-2 levels in the nucleus, modifies the nucleolus, and, potentially as a consequence of a host surveillance mechanism, causes increased defence gene expression. Our results highlight the remarkably complex and potentially antagonistic mechanisms that come into play in the interaction between co-evolved hosts and pathogens.


Subject(s)
Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans/immunology , Enterotoxins/genetics , Hypocreales/pathogenicity , Immunity, Innate , STAT Transcription Factors/genetics , Spores, Fungal/pathogenicity , Animals , Antimicrobial Cationic Peptides/genetics , Antimicrobial Cationic Peptides/immunology , Biological Coevolution , Biological Transport , Caenorhabditis elegans/genetics , Caenorhabditis elegans/metabolism , Caenorhabditis elegans/microbiology , Caenorhabditis elegans Proteins/immunology , Enterotoxins/metabolism , Epidermis/immunology , Epidermis/metabolism , Epidermis/microbiology , Fungal Proteins/genetics , Fungal Proteins/metabolism , Gene Expression Regulation , Genes, Reporter , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Hypocreales/growth & development , Longevity/genetics , Longevity/immunology , STAT Transcription Factors/immunology , Signal Transduction , Spores, Fungal/growth & development , Transport Vesicles/metabolism , Virulence , Virulence Factors/genetics , Virulence Factors/metabolism
14.
J Drugs Dermatol ; 20(2): 199-202, 2021 02 01.
Article in English | MEDLINE | ID: mdl-33538555

ABSTRACT

BACKGROUND: Epidermal inclusion cysts (EIC) are one of the most common forms of cysts found on and/or underneath the skin. Inflamed EICs typically show signs and symptoms such as pain and erythema, mimicking cutaneous abscess. However, prior studies have demonstrated at least 20% of lesions are culture negative. OBJECTIVE: To determine the rate of culture positivity in mild inflamed epidermal inclusion cysts, in particular to identify whether empiric antibiotics are warranted. METHODS: In a retrospective chart review 76 cases of inflamed EIC that were mild (lacking systemic symptoms) were analyzed who presented to the department of dermatology at Mount Sinai between 2016–2019. RESULTS: Of cultures taken from inflamed cysts, 47% resulted in no bacterial growth or growth of normal flora, 38.4% resulted in growth of aerobic bacteria with methicillin-resistant Staphylococcus aureus (8%), Staphylococcus lugdunensis (5%), and methicillin-sensitive Staphylococcus aureus (13%) predominating, and 9.3% resulting in growth of anaerobic bacteria with Finegoldia magna, Peptostreptococcus, and Cutibacterium acnes presenting. Review of prescribed treatment regimens often involved antibiotic medication, despite a high prevalence of negative culture. CONCLUSIONS: Almost half of cases of mild inflamed EIC (lacking systemic symptoms) cultured will not grow pathogenic bacteria, therefore incision and drainage with culture and appropriate therapy is a viable therapeutic option in uncomplicated inflamed EIC lesions. In this way, over prescription of antibiotics can be minimized. J Drugs Dermatol. 2021;20(2):199-202. doi:10.36849/JDD.5014.


Subject(s)
Abscess/diagnosis , Anti-Bacterial Agents/therapeutic use , Drainage , Epidermal Cyst/diagnosis , Gram-Positive Bacterial Infections/diagnosis , Abscess/microbiology , Abscess/therapy , Anti-Bacterial Agents/pharmacology , Clinical Decision-Making , Diagnosis, Differential , Drug Resistance, Bacterial , Epidermal Cyst/immunology , Epidermal Cyst/microbiology , Epidermal Cyst/therapy , Epidermis/microbiology , Epidermis/pathology , Epidermis/surgery , Firmicutes/isolation & purification , Gram-Positive Bacterial Infections/immunology , Gram-Positive Bacterial Infections/microbiology , Gram-Positive Bacterial Infections/therapy , Humans , Peptostreptococcus/isolation & purification , Propionibacterium acnes/isolation & purification , Retrospective Studies , Staphylococcus/isolation & purification
15.
J Cutan Pathol ; 48(4): 486-494, 2021 Apr.
Article in English | MEDLINE | ID: mdl-32965737

ABSTRACT

BACKGROUND: Mycosis fungoides (MF), the commonest primary cutaneous T-cell lymphoma, has classic and variant types which include hypopigmented MF (HMF). Previous studies have identified distinct clinicopathological profiles in HMF. This study aims to objectively compare the clinicopathological features of HMF with non-HMF lesions in order to characterize salient features of HMF. METHODS: This cross-sectional, retrospective study analyzed biopsy specimens of 87 patients with MF. HMF and non-HMF groups were compared using clinical data, immunophenotypic features and scores given for six histopathological features: dermal infiltrate, basilar and superficially extending epidermotropism, Pautrier microabscesses and dermal and epidermotropic lymphocytic atypia. RESULTS: Seventy-six patients had HMF. Presentation in females (59.21%; p = .04) and patch stage (88.16%; p = .01) in HMF were significant, and HMF presented at a younger mean age when compared to non-HMF. Both groups had equal intensity of epidermotropism, with HMF showing milder dermal infiltrates and significantly less dermal atypia. Pautrier microabscesses were significantly commoner in non-HMF (LR 10.76; p < .01). 94.74% of HMF were CD4-/CD8+. CONCLUSION: HMF presents at a lower age and earlier stage with female predominance compared to non-HMF. Because of milder dermal infiltrates, less dermal atypia, and Pautrier microabscesses, the diagnosis of HMF requires correlation with clinical features and careful assessment of epidermotropic cells.


Subject(s)
Epidermis/pathology , Immunophenotyping/methods , Mycosis Fungoides/diagnosis , Mycosis Fungoides/genetics , Skin Neoplasms/pathology , Adolescent , Adult , Biopsy , CD8-Positive T-Lymphocytes/pathology , Child , Child, Preschool , Cross-Sectional Studies , Epidermis/immunology , Epidermis/microbiology , Female , Humans , Infant , Infant, Newborn , Lymphoma, T-Cell, Cutaneous/pathology , Male , Middle Aged , Mycosis Fungoides/pathology , Mycosis Fungoides/ultrastructure , Pigmentation Disorders/pathology , Retrospective Studies , Young Adult
16.
J Dermatol Sci ; 101(1): 14-21, 2021 Jan.
Article in English | MEDLINE | ID: mdl-33218696

ABSTRACT

BACKGROUND: Skin biopsies represent a gold standard in skin immunology and pathology but can cause pain and induce scarring. Non-invasive techniques will facilitate study recruitment of e.g. patients with paediatric atopic dermatitis (AD), hand eczema or facial dermatitis. OBJECTIVE: By RNA sequencing, we examined whether the stratum corneum transcriptome in AD skin can be assessed by tape stripping, as compared to the epidermal transcriptome of AD in skin biopsies. To make the procedure clinically relevant tape strips were stored and shipped at room temperature for up to 3 days. METHODS: Nine adult Caucasian AD patients and three healthy volunteers were included. Tape samples were collected from non-lesional and lesional skin. Biopsies were collected from lesional skin and were split into epidermis and dermis. Total RNA was extracted, and shotgun sequencing was performed. RESULTS: Shotgun sequencing could be performed on skin cells obtained from two consecutive tape strips which had been stored and shipped at room temperature for up to three days. The most prominent differences between the tape strip and biopsy derived transcriptome were due to structural genes, while established molecular markers of AD, including CCL17, CCL22, IL17A and S100A7-S100A9, were also identified in tape strip samples. Furthermore, the tape strip derived transcriptome showed promise in also analysing the skin microbiome. CONCLUSION: Our study shows that the stratum corneum (SC) transcriptome of AD can be assessed by tape stripping the skin, supporting that this method may be central in future skin biomarker research. NCBI GEO data accession: GSE160501.


Subject(s)
Dermatitis, Atopic/genetics , Epidermis/pathology , Transcriptome/genetics , Adult , Biomarkers/analysis , Biopsy/methods , Case-Control Studies , Dermatitis, Atopic/diagnosis , Dermatitis, Atopic/microbiology , Dermatitis, Atopic/pathology , Epidermis/microbiology , Female , Healthy Volunteers , Humans , Male , Microbiota/genetics , Middle Aged , RNA-Seq , Specimen Handling/methods , Time Factors , Young Adult
17.
Sci Rep ; 10(1): 17845, 2020 10 20.
Article in English | MEDLINE | ID: mdl-33082376

ABSTRACT

Staphylococcus aureus (S. aureus) commonly colonizes the human skin and nostrils. However, it is also associated with a wide variety of diseases. S. aureus is frequently isolated from the skin of patients with atopic dermatitis (AD), and is linked to increased disease severity. S. aureus impairs the skin barrier and triggers inflammation through the secretion of various virulence factors. S. aureus secretes phosphatidylinositol-specific phospholipase C (PI-PLC), which hydrolyses phosphatidylinositol and cleaves glycosylphosphatidylinositol-anchored proteins. However, the role of S. aureus PI-PLC in the pathogenesis of skin diseases, including AD, remains unclear. In this study, we sought to determine the role of S. aureus PI-PLC in the pathogenesis of skin diseases. PI-PLC was observed to enhance the invasion and persistence of S. aureus in keratinocytes. Besides, PI-PLC promoted the penetration of S. aureus through the epidermal barrier in a mouse model of AD and the human organotypic epidermal equivalent. Furthermore, the loss of PI-PLC attenuated epidermal hyperplasia and the infiltration of Gr-1+ cells and CD4+ cells induced by S. aureus infection in the mouse model of AD. Collectively, these results indicate that PI-PLC eases the entry of S. aureus into the dermis and aggravates acanthosis and immune cell infiltration in infected skin.


Subject(s)
Epidermis/microbiology , Phosphoinositide Phospholipase C/metabolism , Staphylococcus aureus/physiology , Animals , Dermatitis, Atopic/microbiology , Disease Models, Animal , Gene Knockdown Techniques , Host-Pathogen Interactions , Humans , Keratinocytes/microbiology , Mice
18.
J Fish Dis ; 43(8): 941-953, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32618004

ABSTRACT

This study was carried out to comprehend the pathogenicity of the bacteria in the epidermis of Labeo rohita inoculated with Aeromonas hydrophila. Alterations in the histopathology of the epidermis were examined using scanning electron microscopy, light microscopy and the localization of iNOS and caspase 3 + ve cells by means of immunohistochemical methods. Skin samples obtained from infected fish at different intervals 2, 4, 6, 8 and 10 days showed significant changes in the cellular components of the epidermis. Epithelial cells often appeared hypertrophied with fragmented and loosely arranged microridges, and in the process of exfoliation. Mucous goblet cells increased significantly in density. Club cells showed degenerative changes, often with simultaneous confluence of adjacent cells and release of their contents. Increase in density of iNOS and caspase 3 + ve cells indicates inflammatory response and apoptosis. This study could provide valuable information on the pathogenesis of the disease, and disease outbreaks in farmed fish. Further, it could provide useful guidelines for fish farmers to take preventive measures for the control of the disease.


Subject(s)
Aeromonas hydrophila/physiology , Aeromonas hydrophila/pathogenicity , Carps , Epidermis/pathology , Fish Diseases/pathology , Gram-Negative Bacterial Infections/veterinary , Skin Diseases, Bacterial/veterinary , Animals , Epidermis/microbiology , Epidermis/ultrastructure , Fish Diseases/microbiology , Gram-Negative Bacterial Infections/microbiology , Gram-Negative Bacterial Infections/pathology , Microscopy, Electron, Scanning/veterinary , Skin Diseases, Bacterial/microbiology , Skin Diseases, Bacterial/pathology , Virulence
19.
Mucosal Immunol ; 13(5): 767-776, 2020 09.
Article in English | MEDLINE | ID: mdl-32457449

ABSTRACT

Unlike epidermal Langerhans cells (LCs) that originate from embryonic precursors and are self-renewed locally, mucosal LCs arise and are replaced by circulating bone marrow (BM) precursors throughout life. While the unique lifecycle of epidermal LCs is associated with an age-dependent decrease in their numbers, whether and how aging has an impact on mucosal LCs remains unclear. Focusing on gingival LCs we found that mucosal LCs are reduced with age but exhibit altered morphology with that observed in aged epidermal LCs. The reduction of gingival but not epidermal LCs in aged mice was microbiota-dependent; nevertheless, the impact of the microbiota on gingival LCs was indirect. We next compared the ability of young and aged BM precursors to differentiate to mucosal LCs. Mixed BM chimeras, as well as differentiation cultures, demonstrated that aged BM has intact if not superior capacity to differentiate into LCs than young BM. This was in line with the higher percentages of mucosal LC precursors, pre-DCs, and monocytes, detected in aged BM. These findings suggest that while aging is associated with reduced LC numbers, the niche rather than the origin controls this process in mucosal barriers.


Subject(s)
Cell Differentiation , Cellular Microenvironment/immunology , Langerhans Cells/immunology , Langerhans Cells/metabolism , Mucous Membrane/immunology , Mucous Membrane/metabolism , Age Factors , Aging/physiology , Animals , Biomarkers , Bone Morphogenetic Protein 7/genetics , Bone Morphogenetic Protein 7/metabolism , Cell Differentiation/genetics , Cell Differentiation/immunology , Cellular Microenvironment/genetics , Cellular Senescence/genetics , Cellular Senescence/immunology , Epidermal Cells/immunology , Epidermal Cells/metabolism , Epidermis/immunology , Epidermis/metabolism , Epidermis/microbiology , Gene Expression , Gingiva/immunology , Gingiva/metabolism , Gingiva/microbiology , Immunophenotyping , Langerhans Cells/cytology , Mice , Microbiota , Mucous Membrane/microbiology , Transforming Growth Factor beta1/genetics , Transforming Growth Factor beta1/metabolism
20.
Front Immunol ; 11: 700, 2020.
Article in English | MEDLINE | ID: mdl-32391012

ABSTRACT

Allergic diseases, such as food allergy (FA), atopic dermatitis (AD), and asthma, are heterogeneous inflammatory immune-mediated disorders that currently constitute a public health issue in many developed countries worldwide. The significant increase in the prevalence of allergic diseases reported over the last few years has closely paralleled substantial environmental changes both on a macro and micro scale, which have led to reduced microbial exposure in early life and perturbation of the human microbiome composition. Increasing evidence shows that early life interactions between the human microbiome and the immune cells play a pivotal role in the development of the immune system. Therefore, the process of early colonization by a "healthy" microbiome is emerging as a key determinant of life-long health. In stark contrast, the perturbation of such a process, which results in changes in the host-microbiome biodiversity and metabolic activities, has been associated with greater susceptibility to immune-mediated disorders later in life, including allergic diseases. Here, we outline recent findings on the potential contribution of the microbiome in the gastrointestinal tract, skin, and airways to the development of FA, AD, and asthma. Furthermore, we address how the modulation of the microbiome composition in these different body districts could be a potential strategy for the prevention and treatment of allergic diseases.


Subject(s)
Asthma/immunology , Asthma/microbiology , Dermatitis, Atopic/immunology , Dermatitis, Atopic/microbiology , Food Hypersensitivity/immunology , Food Hypersensitivity/microbiology , Gastrointestinal Microbiome/immunology , Adult , Animals , Child , Epidermis/immunology , Epidermis/microbiology , Humans , Immunity , Respiratory Mucosa/immunology , Respiratory Mucosa/microbiology
SELECTION OF CITATIONS
SEARCH DETAIL
...