Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 2.107
Filtrar
1.
Arch Dermatol Res ; 316(5): 156, 2024 May 11.
Artículo en Inglés | MEDLINE | ID: mdl-38734816

RESUMEN

Atopic dermatitis (AD) is an inflammatory skin disease with intense pruritus, and chronic skin colonization by Staphylococcus aureus. To understand the inflammatory status in AD, we investigated the inflammasome complex, that activates ASC (Apoptosis-associated speck-like protein containing a CARD), caspase-1 and GSDMD (gasdermin-D), and production of IL-1ß and IL-18. We aimed to evaluate the expression of the inflammasome pathway in the skin of adults with AD. Thirty patients with moderate to severe AD and 20 healthy controls were enrolled in the study. We performed the analysis of the inflammasome components NLRP1, NLRP3, AIM-2, IL-1ß, IL-18, Caspase-1, ASC, GSDMD, and CD68 expression (macrophage marker) by immunohistochemistry and immunofluorescence. The main findings included increased expression of NLRP3, NLRP1 and AIM-2 at dermal level of severe AD; augmented IL-18 and IL-1ß expression at epidermis of moderate and severe patients, and in the dermis of severe AD; augmented expression of ASC, caspase-1 and GSDMD in both epidermis and dermis of moderate and severe AD. We detected positive correlation between caspase-1, GSDMD and IL-1ß (epidermis) and caspase-1 (dermis) and AD severity; NLRP3, AIM-2 and IL-1ß, and NLRP3 with IL-18 in the epidermis; ASC, GSDMD and IL-1ß, and NLRP3, AIM-2, caspase-1, and IL-18 in the dermis. We also evidenced the presence of CD68+ macrophages secreting GSDMD, ASC and IL-1ß in moderate and severe AD. Cutaneous macrophages, early detected in moderate AD, have its role in the disease inflammatory mechanisms. Our study indicates a canonical activation pathway of inflammasomes, reinforced by the chronic status of inflammation in AD. The analysis of the inflammasome complex evidenced an imbalance in its regulation, with increased expression of the evaluated components, which is remarkably in severe AD, emphasizing its relevance as potential disease biomarkers and targets for immunomodulatory interventions.


Asunto(s)
Proteínas Adaptadoras de Señalización CARD , Caspasa 1 , Dermatitis Atópica , Inflamasomas , Interleucina-18 , Interleucina-1beta , Péptidos y Proteínas de Señalización Intracelular , Macrófagos , Proteína con Dominio Pirina 3 de la Familia NLR , Proteínas de Unión a Fosfato , Humanos , Inflamasomas/metabolismo , Inflamasomas/inmunología , Proteínas Adaptadoras de Señalización CARD/metabolismo , Dermatitis Atópica/inmunología , Dermatitis Atópica/metabolismo , Dermatitis Atópica/patología , Macrófagos/metabolismo , Macrófagos/inmunología , Interleucina-1beta/metabolismo , Masculino , Femenino , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas de Unión a Fosfato/metabolismo , Adulto , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Interleucina-18/metabolismo , Caspasa 1/metabolismo , Piel/patología , Piel/inmunología , Piel/metabolismo , Índice de Severidad de la Enfermedad , Persona de Mediana Edad , Antígenos de Diferenciación Mielomonocítica/metabolismo , Adulto Joven , Proteínas Reguladoras de la Apoptosis/metabolismo , Antígenos CD/metabolismo , Proteínas NLR/metabolismo , Estudios de Casos y Controles , Epidermis/inmunología , Epidermis/metabolismo , Epidermis/patología , Gasderminas , Molécula CD68 , Proteínas de Unión al ADN
2.
Nature ; 619(7968): 151-159, 2023 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-37344588

RESUMEN

The peripheral T cell repertoire of healthy individuals contains self-reactive T cells1,2. Checkpoint receptors such as PD-1 are thought to enable the induction of peripheral tolerance by deletion or anergy of self-reactive CD8 T cells3-10. However, this model is challenged by the high frequency of immune-related adverse events in patients with cancer who have been treated with checkpoint inhibitors11. Here we developed a mouse model in which skin-specific expression of T cell antigens in the epidermis caused local infiltration of antigen-specific CD8 T cells with an effector gene-expression profile. In this setting, PD-1 enabled the maintenance of skin tolerance by preventing tissue-infiltrating antigen-specific effector CD8 T cells from (1) acquiring a fully functional, pathogenic differentiation state, (2) secreting significant amounts of effector molecules, and (3) gaining access to epidermal antigen-expressing cells. In the absence of PD-1, epidermal antigen-expressing cells were eliminated by antigen-specific CD8 T cells, resulting in local pathology. Transcriptomic analysis of skin biopsies from two patients with cutaneous lichenoid immune-related adverse events showed the presence of clonally expanded effector CD8 T cells in both lesional and non-lesional skin. Thus, our data support a model of peripheral T cell tolerance in which PD-1 allows antigen-specific effector CD8 T cells to co-exist with antigen-expressing cells in tissues without immunopathology.


Asunto(s)
Antígenos , Linfocitos T CD8-positivos , Tolerancia Inmunológica , Receptor de Muerte Celular Programada 1 , Piel , Animales , Humanos , Ratones , Antígenos/inmunología , Biopsia , Linfocitos T CD8-positivos/citología , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Linfocitos T CD8-positivos/patología , Epidermis/inmunología , Epidermis/metabolismo , Perfilación de la Expresión Génica , Liquen Plano/inmunología , Liquen Plano/patología , Receptor de Muerte Celular Programada 1/inmunología , Receptor de Muerte Celular Programada 1/metabolismo , Piel/citología , Piel/inmunología , Piel/metabolismo , Piel/patología
3.
Proc Natl Acad Sci U S A ; 119(25): e2116027119, 2022 06 21.
Artículo en Inglés | MEDLINE | ID: mdl-35704759

RESUMEN

The epidermis is the outermost layer of the skin and the body's primary barrier to external pathogens; however, the early epidermal immune response remains to be mechanistically understood. We show that the chemokine CXCL14, produced by epidermal keratinocytes, exhibits robust circadian fluctuations and initiates innate immunity. Clearance of the skin pathogen Staphylococcus aureus in nocturnal mice was associated with CXCL14 expression, which was high during subjective daytime and low at night. In contrast, in marmosets, a diurnal primate, circadian CXCL14 expression was reversed. Rhythmically expressed CXCL14 binds to S. aureus DNA and induces inflammatory cytokine production by activating Toll-like receptor (TLR)9-dependent innate pathways in dendritic cells and macrophages underneath the epidermis. CXCL14 also promoted phagocytosis by macrophages in a TLR9-independent manner. These data indicate that circadian production of the epidermal chemokine CXCL14 rhythmically suppresses skin bacterial proliferation in mammals by activating the innate immune system.


Asunto(s)
Epidermis , Inmunidad Innata , Enfermedades Cutáneas Bacterianas , Animales , Quimiocinas CXC/genética , Quimiocinas CXC/inmunología , Relojes Circadianos/inmunología , Epidermis/inmunología , Inmunidad Innata/genética , Inmunidad Innata/inmunología , Queratinocitos/inmunología , Mamíferos , Ratones , Enfermedades Cutáneas Bacterianas/inmunología , Enfermedades Cutáneas Bacterianas/metabolismo , Infecciones Estafilocócicas/inmunología , Staphylococcus aureus/inmunología
4.
Mar Drugs ; 20(3)2022 Feb 22.
Artículo en Inglés | MEDLINE | ID: mdl-35323456

RESUMEN

Crustin are a family of antimicrobial peptides that play an important role in protecting against pathogens infection in the innate immune system of crustaceans. Previously, we identified several novel types of crustins, including type VI and type VII crustins. However, their immune functions were still unclear. In the present study, the immune function of type VII crustin LvCrustinVII were investigated in Litopenaeus vannamei. LvCrustinVII was wildly expressed in all tested tissues, with relatively high expression levels in hepatopancreas, epidermis and lymphoid organ. Upon Vibrio parahaemolyticus infection, LvCrustinVII was significantly upregulated in hepatopancreas. Recombinant LvCrustinVII (rLvCrustinVII) showed strong inhibitory activities against Gram-negative bacteria Vibrio harveyi and V. parahaemolyticus, while weak activities against the Gram-positive bacteria Staphylococcus aureus. Binding assay showed that rLvCrustinVII could bind strongly to V. harveyi and V. parahaemolyticus, as well as the cell wall components Glu, LPS and PGN. In the presence of Ca2+, rLvCrustinVII could agglutinate V. parahaemolyticus and enhance hemocyte phagocytosis. The present data partially illustrate the immune function of LvCrustinVII, which enrich our understanding on the functional mechanisms of crustins and provide useful information for application of this kind of antimicrobial peptides.


Asunto(s)
Péptidos Catiónicos Antimicrobianos , Proteínas de Artrópodos , Proteínas Opsoninas , Penaeidae/inmunología , Aglutinación , Animales , Péptidos Catiónicos Antimicrobianos/química , Péptidos Catiónicos Antimicrobianos/genética , Péptidos Catiónicos Antimicrobianos/inmunología , Péptidos Catiónicos Antimicrobianos/farmacología , Proteínas de Artrópodos/química , Proteínas de Artrópodos/genética , Proteínas de Artrópodos/inmunología , Proteínas de Artrópodos/farmacología , Bacterias/química , Bacterias/efectos de los fármacos , Bacterias/genética , Bacterias/crecimiento & desarrollo , Epidermis/inmunología , Hemocitos/fisiología , Hepatopáncreas/inmunología , Proteínas Opsoninas/química , Proteínas Opsoninas/genética , Proteínas Opsoninas/inmunología , Proteínas Opsoninas/farmacología , Fagocitosis , Proteínas Recombinantes/química , Proteínas Recombinantes/farmacología
5.
J Invest Dermatol ; 142(2): 402-413, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34333017

RESUMEN

Dermal fibroblasts are strategically positioned underneath the basal epidermis layer to support keratinocyte proliferation and extracellular matrix production. In inflammatory conditions, these fibroblasts produce cytokines and chemokines that promote the chemoattraction of immune cells into the dermis and the hyperplasia of the epidermis, two characteristic hallmarks of psoriasis. However, how dermal fibroblasts specifically contribute to psoriasis development remains largely uncharacterized. In this study, we investigated through which cytokines and signaling pathways dermal fibroblasts contribute to the inflammatory features of psoriatic skin. We show that dermal fibroblasts from lesional psoriatic skin are important producers of inflammatory mediators, including IL-6, CXCL8, and CXCL2. This increased cytokine production was found to be regulated by ZFP36 family members ZFP36, ZFP36L1, and ZFP36L2, RNA-binding proteins with mRNA-degrading properties. In addition, the expression of ZFP36 family proteins was found to be reduced in chronic inflammatory conditions that mimic psoriatic lesional skin. Collectively, these results indicate that dermal fibroblasts are important producers of cytokines in psoriatic skin and that reduced expression of ZFP36 members in psoriasis dermal fibroblasts contributes to their inflammatory phenotype.


Asunto(s)
Factor 1 de Respuesta al Butirato/metabolismo , Fibroblastos/metabolismo , Psoriasis/inmunología , Factores de Transcripción/metabolismo , Tristetraprolina/metabolismo , Biopsia , Factor 1 de Respuesta al Butirato/genética , Estudios de Casos y Controles , Epidermis/inmunología , Epidermis/metabolismo , Epidermis/patología , Técnicas de Silenciamiento del Gen , Voluntarios Sanos , Humanos , Mediadores de Inflamación/metabolismo , Queratinocitos/inmunología , Queratinocitos/metabolismo , Psoriasis/patología , Factores de Transcripción/genética , Tristetraprolina/genética
6.
Allergol Int ; 71(1): 3-13, 2022 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-34916117

RESUMEN

Atopic dermatitis (AD) is the most common chronic skin inflammatory disease, with a profound impact on patients' quality of life. AD varies considerably in clinical course, age of onset and degree to which it is accompanied by allergic and non-allergic comorbidities. Skin barrier impairment in both lesional and nonlesional skin is now recognized as a critical and often early feature of AD. This may be explained by a number of abnormalities identified within both the stratum corneum and stratum granulosum layers of the epidermis. The goal of this review is to provide an overview of key barrier defects in AD, starting with a historical perspective. We will also highlight some of the commonly used methods to characterize and quantify skin barrier function. There is ample opportunity for further investigative work which we call out throughout this review. These include: quantifying the relative impact of individual epidermal abnormalities and putting this in a more holistic view with physiological measures of barrier function, as well as determining whether these barrier-specific endotypes predict clinical phenotypes (e.g. age of onset, natural history, comorbidities, response to therapies, etc). Mechanistic studies with new (and in development) AD therapies that specifically target immune pathways, Staphylococcus aureus abundance and/or skin barrier will help us understand the dynamic crosstalk between these compartments and their relative importance in AD.


Asunto(s)
Dermatitis Atópica/inmunología , Epidermis/inmunología , Animales , Progresión de la Enfermedad , Humanos , Calidad de Vida , Índice de Severidad de la Enfermedad
7.
Front Immunol ; 12: 775412, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34899732

RESUMEN

Laminin-332 pemphigoid is a rare and severe autoimmune blistering disease, caused by IgG autoantibodies targeting laminin-332 in the dermal-epidermal basement zone. Laminin-332 pemphigoid is characterized by variable inflammatory infiltrate and the predominance of non-complement-fixing antibodies. Given these findings, we hypothesized that IgG autoantibodies to laminin-332 directly resulted in keratinocyte expression of inflammatory factors. We performed RNA-seq on primary human keratinocytes treated with IgG from patients with laminin-332 pemphigoid. Genes for numerous cytokines and chemokines were upregulated, including CSF2, CSF3, CXCL1, CXCL5, CXCL3, CXCL8, CXCL10, CXCL1, IL6, IL7, IL15, IL23, IL32, IL37, TGFB2 as well as metalloproteases. Considering the pro-inflammatory and proteolytic effect of autoantibodies from patients with laminin-332 pemphigoid identified in our initial experiment, we next questioned whether the reactivity against specific laminin subunits dictates the inflammatory and proteolytic keratinocyte response. Then, we treated keratinocytes with IgG from a separate cohort of patients with reactivity against individual subunits of laminin-332. We identified upregulation of IL-1α, IL-6, IL-8, CXCL1, MMP9, TSLP, and GM-CSF at the protein level, most notably in keratinocytes treated with IgG from laminin ß3-reactive patients. We for the first time demonstrated a pro-inflammatory response, similar to that described in keratinocytes treated with IgG autoantibodies from patients with bullous pemphigoid, providing novel insight into the pathogenesis of laminin-332 pemphigoid and laminin-332 biology.


Asunto(s)
Autoanticuerpos/metabolismo , Autoantígenos/inmunología , Moléculas de Adhesión Celular/inmunología , Citocinas/metabolismo , Epidermis/metabolismo , Inmunoglobulina G/metabolismo , Mediadores de Inflamación/metabolismo , Queratinocitos/metabolismo , Penfigoide Benigno de la Membrana Mucosa/metabolismo , Anciano , Anciano de 80 o más Años , Especificidad de Anticuerpos , Células Cultivadas , Citocinas/genética , Epidermis/inmunología , Femenino , Perfilación de la Expresión Génica , Humanos , Queratinocitos/inmunología , Masculino , Persona de Mediana Edad , Penfigoide Benigno de la Membrana Mucosa/inmunología , RNA-Seq , Transcriptoma , Kalinina
8.
Front Immunol ; 12: 774018, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34925344

RESUMEN

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.


Asunto(s)
Infecciones Bacterianas/transmisión , Epidermis/microbiología , Bacterias/crecimiento & desarrollo , Epidermis/inmunología , Dedos/microbiología , Mano/microbiología , Humanos , Microbiota
9.
J Clin Invest ; 131(24)2021 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-34730110

RESUMEN

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.


Asunto(s)
Pie Diabético/inmunología , Epidermis/inmunología , Proteínas de la Membrana/inmunología , Proteínas Citotóxicas Formadoras de Poros/inmunología , Piroptosis/inmunología , Infecciones Estafilocócicas/inmunología , Staphylococcus aureus/inmunología , Cicatrización de Heridas/inmunología , Adulto , Anciano , Animales , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/inmunología , Pie Diabético/genética , Pie Diabético/microbiología , Epidermis/microbiología , Femenino , Humanos , Interleucina-1beta/genética , Interleucina-1beta/inmunología , Masculino , Proteínas de la Membrana/genética , Ratones , Ratones Noqueados , Persona de Mediana Edad , Proteínas Citotóxicas Formadoras de Poros/genética , Piroptosis/genética , Infecciones Estafilocócicas/genética , Cicatrización de Heridas/genética
10.
Immunohorizons ; 5(10): 830-843, 2021 10 26.
Artículo en Inglés | MEDLINE | ID: mdl-34702760

RESUMEN

Atopic dermatitis (AD) is a chronic inflammatory skin disease that affects up to one in five children and millions of adults in developed countries. Clinically, AD skin lesions manifest as subacute and/or chronic lichenified eczematous plaques, which are often intensely pruritic and prone to secondary bacterial and viral infections. Despite the emergence of novel therapeutic agents, treatment options and outcomes for AD remain suboptimal. An improved understanding of AD pathogenesis may help improve patient outcomes. Dysregulated Th2-polarized skin inflammation and impaired skin barrier function interact to drive AD pathogenesis; however, much remains to be understood about the molecular mechanisms underlying this interplay. The current study used published clinical trial datasets to define a skin-related AD gene signature. This meta-analysis revealed significant reductions in IL1F7 transcripts (encodes IL-37) in AD patient samples. Reduced IL1F7 correlated with lower transcripts for key skin barrier function genes in the epidermal differentiation complex. Immunohistochemical analysis of normal (healthy) human skin specimens and an in vitro three-dimensional human skin model localized IL-37 protein to the epidermis. In comparison with normal human skin, IL-37 levels were decreased in AD patient skin. Addition of Th2 cytokines to the aforementioned in vitro three-dimensional skin model recapitulates key aspects of AD skin and was sufficient to reduce epidermal IL-37 levels. Image analysis also indicated close relationship between epidermal IL-37 and skin epidermal differentiation complex proteins. These findings suggest IL-37 is intimately linked to normal keratinocyte differentiation and barrier function and implicates IL-37 as a potential biomarker and therapeutic target for AD.


Asunto(s)
Dermatitis Atópica/inmunología , Epidermis/patología , Interleucina-1/metabolismo , Adulto , Azetidinas/uso terapéutico , Biopsia , Diferenciación Celular/inmunología , Dermatitis Atópica/diagnóstico , Dermatitis Atópica/tratamiento farmacológico , Dermatitis Atópica/patología , Regulación hacia Abajo/inmunología , Epidermis/inmunología , Epidermis/metabolismo , Femenino , Perfilación de la Expresión Génica , Voluntarios Sanos , Humanos , Queratinocitos/inmunología , Queratinocitos/metabolismo , Masculino , Persona de Mediana Edad , Purinas/uso terapéutico , Pirazoles/uso terapéutico , Índice de Severidad de la Enfermedad , Sulfonamidas/uso terapéutico , Células Th2/inmunología , Células Th2/metabolismo
11.
Front Immunol ; 12: 695373, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34512625

RESUMEN

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.


Asunto(s)
Péptidos Antimicrobianos/genética , Epidermis/metabolismo , Células de Langerhans/metabolismo , Infecciones Cutáneas Estafilocócicas/genética , Staphylococcus aureus/patogenicidad , Transcriptoma , Células Cultivadas , Bases de Datos Genéticas , Epidermis/inmunología , Epidermis/microbiología , Perfilación de la Expresión Génica , Interacciones Huésped-Patógeno , Humanos , Células de Langerhans/inmunología , Células de Langerhans/microbiología , Infecciones Cutáneas Estafilocócicas/inmunología , Infecciones Cutáneas Estafilocócicas/metabolismo , Infecciones Cutáneas Estafilocócicas/microbiología , Staphylococcus aureus/inmunología
12.
Virulence ; 12(1): 2474-2492, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34516337

RESUMEN

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.


Asunto(s)
Toxinas Bacterianas/metabolismo , Epidermis , Secretoma , Infecciones Estafilocócicas , Staphylococcus aureus , Factores de Virulencia , Quimiocinas/inmunología , Citocinas/inmunología , Epidermis/inmunología , Epidermis/microbiología , Humanos , Inflamación , Mediadores de Inflamación/inmunología , Staphylococcus aureus/metabolismo , Factores de Virulencia/metabolismo
13.
Sci Rep ; 11(1): 16091, 2021 08 09.
Artículo en Inglés | MEDLINE | ID: mdl-34373514

RESUMEN

Cold atmospheric plasma (CAP) has been incorporated into various fields, including promotion of cutaneous wound healing. Atopic dermatitis (AD) is a chronic cutaneous condition characterized by inflammation-induced skin wounds and impaired skin barrier function. To investigate whether CAP may improve AD using an animal model. Dermatophagoides farinae extracts (DFE)-induced murine models of AD were used in this study. The plasma-treated group received a total of 6 CAP treatments during 2 weeks, while the control group did not receive any treatment. Differences in dermatitis severity, transepidermal water loss (TEWL), serum level of immunoglobulin (Ig) E and epidermal thickness were evaluated in both groups. The dermatitis severity was significantly improved by CAP treatment. TEWL was lower in the plasma-treated group compared with the non-treated control group. Serum Ig E dropped significantly after treatment with CAP. Difference in epidermal thickness of the ear skin was not significant between the plasma-treated and non-treated groups. Localized treatment of AD with CAP decreases dermatitis severity, TEWL, and serum Ig E level. These results show CAP's potentials as a novel therapeutic modality for AD.


Asunto(s)
Dermatitis Atópica/inmunología , Dermatitis Atópica/terapia , Animales , Dermatophagoides farinae/inmunología , Modelos Animales de Enfermedad , Epidermis/inmunología , Inmunoglobulina E/inmunología , Inflamación/inmunología , Inflamación/terapia , Masculino , Ratones , Piel/inmunología
14.
Eur J Immunol ; 51(11): 2618-2632, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34398456

RESUMEN

The epidermis of mouse skin is usually populated by dendritic epidermal T cells (γδDETC) expressing an invariant Vγ5Vδ1+ TCR. In Tcrd-/- mice, skin-resident γδDETC are replaced by αßDETC carrying polyclonal αß TCRs. Although they exhibit a dendritic morphology, αßDETC were reported to be less functional than genuine γδDETC, likely because their TCR is unable to interact with the original TCR ligands of γδDETC. However, the TCR repertoire of those replacement DETC in Tcrd-/- mice might provide clues for understanding the development and selection of canonical γδDETC. Here, we compare the phenotype and TCR repertoires of wild-type and Tcrd-/- mouse skin T cells. Our data reveal that αßDETC are CD4/CD8 double negative and express an MHC-independent TCR repertoire. Furthermore, we identify a second MHC-independent population of CD103hi CD4/ CD8 double-negative αß T cells in the dermis of Tcrd-/- mice.


Asunto(s)
Células Dendríticas/inmunología , Células Epidérmicas/inmunología , Receptores de Antígenos de Linfocitos T alfa-beta/inmunología , Receptores de Antígenos de Linfocitos T gamma-delta/inmunología , Subgrupos de Linfocitos T/inmunología , Animales , Epidermis/inmunología , Ratones , Ratones Noqueados
15.
Molecules ; 26(15)2021 Jul 21.
Artículo en Inglés | MEDLINE | ID: mdl-34361560

RESUMEN

The extracts of Schisandra chinensis (Turcz.) Baill. (Schisandraceae) have various therapeutic effects, including inflammation and allergy. In this study, gomisin M2 (GM2) was isolated from S. chinensis and its beneficial effects were assessed against atopic dermatitis (AD). We evaluated the therapeutic effects of GM2 on 2,4-dinitrochlorobenzene (DNCB) and Dermatophagoides farinae extract (DFE)-induced AD-like skin lesions with BALB/c mice ears and within the tumor necrosis factor (TNF)-α and interferon (IFN)-γ-stimulated keratinocytes. The oral administration of GM2 resulted in reduced epidermal and dermal thickness, infiltration of tissue eosinophils, mast cells, and helper T cells in AD-like lesions. GM2 suppressed the expression of IL-1ß, IL-4, IL-5, IL-6, IL-12a, and TSLP in ear tissue and the expression of IFN-γ, IL-4, and IL-17A in auricular lymph nodes. GM2 also inhibited STAT1 and NF-κB phosphorylation in DNCB/DFE-induced AD-like lesions. The oral administration of GM2 reduced levels of IgE (DFE-specific and total) and IgG2a in the mice sera, as well as protein levels of IL-4, IL-6, and TSLP in ear tissues. In TNF-α/IFN-γ-stimulated keratinocytes, GM2 significantly inhibited IL-1ß, IL-6, CXCL8, and CCL22 through the suppression of STAT1 phosphorylation and the nuclear translocation of NF-κB. Taken together, these results indicate that GM2 is a biologically active compound that exhibits inhibitory effects on skin inflammation and suggests that GM2 might serve as a remedy in inflammatory skin diseases, specifically on AD.


Asunto(s)
Antiinflamatorios/farmacología , Ciclooctanos/farmacología , Dermatitis Atópica , Dermatophagoides farinae/inmunología , Dermis/inmunología , Dinitroclorobenceno/toxicidad , Epidermis/inmunología , FN-kappa B/inmunología , Factor de Transcripción STAT1/inmunología , Animales , Antiinflamatorios/química , Ciclooctanos/química , Citocinas/inmunología , Dermatitis Atópica/inducido químicamente , Dermatitis Atópica/tratamiento farmacológico , Dermatitis Atópica/inmunología , Ratones , Ratones Endogámicos BALB C
16.
Int J Mol Sci ; 22(16)2021 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-34445108

RESUMEN

Atopic dermatitis (AD) is a common inflammatory dermatosis affecting up to 30% of children and 10% of adults worldwide. AD is primarily driven by an epidermal barrier defect which triggers immune dysregulation within the skin. According to recent research such phenomena are closely related to the microbial dysbiosis of the skin. There is growing evidence that cutaneous microbiota and bacterial biofilms negatively affect skin barrier function, contributing to the onset and exacerbation of AD. This review summarizes the latest data on the mechanisms leading to microbiome dysbiosis and biofilm formation in AD, and the influence of these phenomena on skin barrier function.


Asunto(s)
Bacterias/inmunología , Biopelículas/crecimiento & desarrollo , Dermatitis Atópica/inmunología , Disbiosis/inmunología , Epidermis/inmunología , Microbiota/inmunología , Animales , Disbiosis/microbiología , Humanos , Piel/inmunología
17.
Cells ; 10(6)2021 06 04.
Artículo en Inglés | MEDLINE | ID: mdl-34200009

RESUMEN

Atopic dermatitis (AD) is one of the most prevalent inflammatory disease among non-fatal skin diseases, affecting up to one fifth of the population in developed countries. AD is characterized by recurrent pruritic and localized eczema with seasonal fluctuations. AD initializes the phenomenon of atopic march, during which infant AD patients are predisposed to progressive secondary allergies such as allergic rhinitis, asthma, and food allergies. The pathophysiology of AD is complex; onset of the disease is caused by several factors, including strong genetic predisposition, disrupted epidermal barrier, and immune dysregulation. AD was initially characterized by defects in the innate immune system and a vigorous skewed adaptive Th2 response to environmental agents; there are compelling evidences that the disorder involves multiple immune pathways. Symptomatic palliative treatment is the only strategy to manage the disease and restore skin integrity. Researchers are trying to more precisely define the contribution of different AD genotypes and elucidate the role of various immune axes. In this review, we have summarized the current knowledge about the roles of innate and adaptive immune responsive cells in AD. In addition, current and novel treatment strategies for the management of AD are comprehensively described, including some ongoing clinical trials and promising therapeutic agents. This information will provide an asset towards identifying personalized targets for better therapeutic outcomes.


Asunto(s)
Inmunidad Adaptativa , Dermatitis Atópica/inmunología , Epidermis/inmunología , Inmunidad Innata , Células Th2/inmunología , Animales , Asma/etiología , Asma/inmunología , Asma/patología , Dermatitis Atópica/complicaciones , Dermatitis Atópica/patología , Epidermis/patología , Hipersensibilidad a los Alimentos/etiología , Hipersensibilidad a los Alimentos/inmunología , Hipersensibilidad a los Alimentos/patología , Humanos , Rinitis Alérgica/etiología , Rinitis Alérgica/inmunología , Rinitis Alérgica/patología , Células Th2/patología
18.
PLoS Genet ; 17(6): e1009600, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-34166401

RESUMEN

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.


Asunto(s)
Proteínas de Caenorhabditis elegans/genética , Caenorhabditis elegans/inmunología , Enterotoxinas/genética , Hypocreales/patogenicidad , Inmunidad Innata , Factores de Transcripción STAT/genética , Esporas Fúngicas/patogenicidad , Animales , Péptidos Catiónicos Antimicrobianos/genética , Péptidos Catiónicos Antimicrobianos/inmunología , Coevolución Biológica , Transporte Biológico , Caenorhabditis elegans/genética , Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/microbiología , Proteínas de Caenorhabditis elegans/inmunología , Enterotoxinas/metabolismo , Epidermis/inmunología , Epidermis/metabolismo , Epidermis/microbiología , Proteínas Fúngicas/genética , Proteínas Fúngicas/metabolismo , Regulación de la Expresión Génica , Genes Reporteros , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Interacciones Huésped-Patógeno/genética , Interacciones Huésped-Patógeno/inmunología , Hypocreales/crecimiento & desarrollo , Longevidad/genética , Longevidad/inmunología , Factores de Transcripción STAT/inmunología , Transducción de Señal , Esporas Fúngicas/crecimiento & desarrollo , Vesículas Transportadoras/metabolismo , Virulencia , Factores de Virulencia/genética , Factores de Virulencia/metabolismo
19.
J Dermatol Sci ; 102(3): 142-157, 2021 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-34116898

RESUMEN

Atopic dermatitis (AD) is a chronic, inflammatory skin disorder characterized by eczematous and pruritic skin lesions. In recent decades, the prevalence of AD has increased worldwide, most notably in developing countries. The enormous progress in our understanding of the complex composition and functions of the epidermal barrier allows for a deeper appreciation of the active role that the skin barrier plays in the initiation and maintenance of skin inflammation. The epidermis forms a physical, chemical, immunological, neuro-sensory, and microbial barrier between the internal and external environment. Not only lesional, but also non-lesional areas of AD skin display many morphological, biochemical and functional differences compared with healthy skin. Supporting this notion, genetic defects affecting structural proteins of the skin barrier, including filaggrin, contribute to an increased risk of AD. There is evidence to suggest that natural environmental allergens and man-made pollutants are associated with an increased likelihood of developing AD. A compromised epidermal barrier predisposes the skin to increased permeability of these compounds. Numerous topical and systemic therapies for AD are currently available or in development; while anti-inflammatory therapy is central to the treatment of AD, some existing and novel therapies also appear to exert beneficial effects on skin barrier function. Further research on the skin barrier, particularly addressing epidermal differentiation and inflammation, lipid metabolism, and the role of bacterial communities for skin barrier function, will likely expand our understanding of the complex etiology of AD and lead to identification of novel targets and the development of new therapies.


Asunto(s)
Dermatitis Atópica/inmunología , Fármacos Dermatológicos/farmacocinética , Epidermis/patología , Microbiota/inmunología , Diferenciación Celular/efectos de los fármacos , Dermatitis Atópica/tratamiento farmacológico , Dermatitis Atópica/patología , Fármacos Dermatológicos/uso terapéutico , Desarrollo de Medicamentos , Epidermis/efectos de los fármacos , Epidermis/inmunología , Proteínas Filagrina , Humanos , Metabolismo de los Lípidos/efectos de los fármacos , Metabolismo de los Lípidos/inmunología , Microbiota/efectos de los fármacos , Permeabilidad/efectos de los fármacos , Pérdida Insensible de Agua/efectos de los fármacos , Pérdida Insensible de Agua/inmunología
20.
Nat Cell Biol ; 23(5): 476-484, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33958758

RESUMEN

Organs consist of multiple cell types that ensure proper architecture and function. How different cell types coexist and interact to maintain their homeostasis in vivo remains elusive. The skin epidermis comprises mostly epithelial cells, but also harbours Langerhans cells (LCs) and dendritic epidermal T cells (DETCs). Whether and how distributions of LCs and DETCs are regulated during homeostasis is unclear. Here, by tracking individual cells in the skin of live adult mice over time, we show that LCs and DETCs actively maintain a non-random spatial distribution despite continuous turnover of neighbouring basal epithelial cells. Moreover, the density of epithelial cells regulates the composition of LCs and DETCs in the epidermis. Finally, LCs require the GTPase Rac1 to maintain their positional stability, density and tiling pattern reminiscent of neuronal self-avoidance. We propose that these cellular mechanisms provide the epidermis with an optimal response to environmental insults.


Asunto(s)
Células Epidérmicas/citología , Epidermis/metabolismo , Piel/citología , Linfocitos T/inmunología , Animales , Células Epidérmicas/inmunología , Epidermis/inmunología , Homeostasis/inmunología , Homeostasis/fisiología , Uniones Intercelulares/patología , Ratones Transgénicos , Piel/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...