Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 1.831
Filtrer
1.
Int J Mol Sci ; 25(11)2024 Jun 03.
Article de Anglais | MEDLINE | ID: mdl-38892346

RÉSUMÉ

Atopic dermatitis (AD) is one of the most common chronic inflammatory skin diseases, with an increasing number of targeted therapies available. While biologics to treat AD exclusively target the key cytokines of type 2 immunity, Janus kinase inhibitors target a broad variety of cytokines, including IFN-γ. To better stratify patients for optimal treatment outcomes, the identification and characterization of subgroups, especially with regard to their IFNG expression, is of great relevance, as the role of IFNG in AD has not yet been fully clarified. This study aims to define AD subgroups based on their lesional IFNG expression and to characterize them based on their gene expression, T cell secretome and clinical attributes. RNA from the lesional and non-lesional biopsies of 48 AD patients was analyzed by RNA sequencing. Based on IFNG gene expression and the release of IFN-γ by lesional T cells, this cohort was categorized into three IFNG groups (high, medium, and low) using unsupervised clustering. The low IFNG group showed features of extrinsic AD with a higher prevalence of atopic comorbidities and impaired epidermal lipid synthesis. In contrast, patients in the high IFNG group had a higher average age and an activation of additional pro-inflammatory pathways. On the cellular level, higher amounts of M1 macrophages and natural killer cell signaling were detected in the high IFNG group compared to the low IFNG group by a deconvolution algorithm. However, both groups shared a common dupilumab response gene signature, indicating that type 2 immunity is the dominant immune shift in both subgroups. In summary, high and low IFNG subgroups correspond to intrinsic and extrinsic AD classifications and might be considered in the future for evaluating therapeutic efficacy or non-responders.


Sujet(s)
Eczéma atopique , Interféron gamma , Eczéma atopique/génétique , Eczéma atopique/métabolisme , Eczéma atopique/immunologie , Humains , Interféron gamma/métabolisme , Interféron gamma/génétique , Femelle , Mâle , Adulte , Adulte d'âge moyen , Anticorps monoclonaux humanisés/usage thérapeutique , Macrophages/métabolisme , Macrophages/immunologie , Lymphocytes T/métabolisme , Lymphocytes T/immunologie , Cellules tueuses naturelles/métabolisme , Cellules tueuses naturelles/immunologie
2.
Int J Mol Sci ; 25(11)2024 May 24.
Article de Anglais | MEDLINE | ID: mdl-38891925

RÉSUMÉ

Stress exposure worsens allergic inflammatory diseases substantially. Mast cells (MCs) play a key role in peripheral immune responses to neuroendocrine stress mediators such as nerve growth factor (NGF) and substance P (SP). Mast cell proteases (MCPs) and cholinergic factors (Chrna7, SLURP1) were recently described to modulate MC stress response. We studied MCPs and Chrna7/SLURP1 and their interplay in a mouse model for noise induced stress (NiS) and atopic dermatitis-like allergic inflammation (AlD) and in cultured MC lacking Chrna7. We found that the cholinergic stress axis interacts with neuroendocrine stress mediators and stress-mediator cleaving enzymes in AlD. SP-cleaving mMCP4+ MC were upregulated in AlD and further upregulated by stress in NiS+AlD. Anti-NGF neutralizing antibody treatment blocked the stress-induced upregulation in vivo, and mMCP4+ MCs correlated with measures of AlD disease activity. Finally, high mMCP4 production in response to SP depended on Chrna7/SLURP1 in cultured MCs. In conclusion, mMCP4 and its upstream regulation by Chrna7/SLURP1 are interesting novel targets for the treatment of allergic inflammation and its aggravation by stress.


Sujet(s)
Eczéma atopique , Modèles animaux de maladie humaine , Mastocytes , Peau , Récepteur nicotinique de l'acétylcholine alpha7 , Animaux , Mastocytes/métabolisme , Mastocytes/immunologie , Eczéma atopique/métabolisme , Eczéma atopique/anatomopathologie , Eczéma atopique/immunologie , Souris , Peau/métabolisme , Peau/anatomopathologie , Récepteur nicotinique de l'acétylcholine alpha7/métabolisme , Inflammation/métabolisme , Inflammation/anatomopathologie , Peptide hydrolases/métabolisme , Activateur du plasminogène de type urokinase/métabolisme , Substance P/métabolisme , Stress physiologique , Souris de lignée C57BL , Facteur de croissance nerveuse/métabolisme
3.
Int J Biol Sci ; 20(8): 3094-3112, 2024.
Article de Anglais | MEDLINE | ID: mdl-38904012

RÉSUMÉ

Atopic dermatitis (AD) is a common inflammation skin disease that involves dysregulated interplay between immune cells and keratinocytes. Interleukin-38 (IL-38), a poorly characterized IL-1 family cytokine, its role and mechanism in the pathogenesis of AD is elusive. Here, we show that IL-38 is mainly secreted by epidermal keratinocytes and highly expressed in the skin and downregulated in AD lesions. We generated IL-38 keratinocyte-specific knockout mice (K14Cre/+-IL-38f/f ) and induced AD models by 2,4-dinitrofluorobenzene (DNFB). Unexpectedly, after treatment with DNFB, K14Cre/+-IL-38f/f mice were less susceptible to cutaneous inflammation of AD. Moreover, keratinocyte-specific deletion of IL-38 suppressed the migration of Langerhans cells (LCs) into lymph nodes which results in disturbed differentiation of CD4+T cells and decreased the infiltration of immune cells into AD lesions. LCs are a type of dendritic cell that reside specifically in the epidermis and regulate immune responses. We developed LC-like cells in vitro from mouse bone marrow (BM) and treated with recombined IL-38. The results show that IL-38 depended on IL-36R, activated the phosphorylated expression of IRAK4 and NF-κB P65 and upregulated the expression of CCR7 to promoting the migration of LCs, nevertheless, the upregulation disappeared with the addition of IL-36 receptor antagonist (IL-36RA), IRAK4 or NF-κB P65 inhibitor. Furthermore, after treatment with IRAK4 inhibitors, the experimental AD phenotypes were alleviated and so IRAK4 is considered a promising target for the treatment of inflammatory diseases. Overall, our findings indicated a potential pathway that IL-38 depends on IL-36R, leading to LCs migration to promote AD by upregulating CCR7 via IRAK4/NF-κB and implied the prevention and treatment of AD, supporting potential clinical utilization of IRAK4 inhibitors in AD treatment.


Sujet(s)
Mouvement cellulaire , Eczéma atopique , Cellules de Langerhans , Animaux , Eczéma atopique/métabolisme , Cellules de Langerhans/métabolisme , Souris , Souris knockout , Interleukine-1/métabolisme , Kératinocytes/métabolisme , 1-Fluoro-2,4-dinitro-benzène , Facteur de transcription NF-kappa B/métabolisme , Interleukines/métabolisme
4.
Arch Dermatol Res ; 316(5): 156, 2024 May 11.
Article de Anglais | MEDLINE | ID: mdl-38734816

RÉSUMÉ

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.


Sujet(s)
Protéines adaptatrices de signalisation CARD , Caspase-1 , Eczéma atopique , Inflammasomes , Macrophages , Protéine-3 de la famille des NLR contenant un domaine pyrine , Adulte , Femelle , Humains , Mâle , Adulte d'âge moyen , Jeune adulte , Antigènes CD/métabolisme , Antigènes de différenciation des myélomonocytes/métabolisme , Protéines régulatrices de l'apoptose/métabolisme , Protéines adaptatrices de signalisation CARD/métabolisme , Études cas-témoins , Caspase-1/métabolisme , , Eczéma atopique/immunologie , Eczéma atopique/métabolisme , Eczéma atopique/anatomopathologie , Protéines de liaison à l'ADN , Épiderme/immunologie , Épiderme/métabolisme , Épiderme/anatomopathologie , Gasdermines , Inflammasomes/métabolisme , Inflammasomes/immunologie , Interleukine-18/métabolisme , Interleukine-1 bêta/métabolisme , Protéines et peptides de signalisation intracellulaire/métabolisme , Macrophages/métabolisme , Macrophages/immunologie , Protéine-3 de la famille des NLR contenant un domaine pyrine/métabolisme , Protéines NLR/métabolisme , Protéines de liaison aux phosphates/métabolisme , Indice de gravité de la maladie , Peau/anatomopathologie , Peau/immunologie , Peau/métabolisme
6.
Int J Mol Sci ; 25(10)2024 May 10.
Article de Anglais | MEDLINE | ID: mdl-38791249

RÉSUMÉ

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


Sujet(s)
Eczéma atopique , Métabolome , Microbiote , Peau , Eczéma atopique/microbiologie , Eczéma atopique/métabolisme , Humains , Peau/microbiologie , Peau/métabolisme , Femelle , Mâle , Adulte , ARN ribosomique 16S/génétique , Métabolomique/méthodes , Jeune adulte , Adulte d'âge moyen , Études cas-témoins
7.
PLoS One ; 19(5): e0302781, 2024.
Article de Anglais | MEDLINE | ID: mdl-38713650

RÉSUMÉ

Atopic dermatitis is a multi-pathogenic disease characterized by chronic skin inflammation and barrier dysfunction. Therefore, improving the skin's ability to form an epidermal barrier and suppressing the production of cytokines that induce type 2 inflammatory responses are important for controlling atopic dermatitis symptoms. (-)-Blebbistatin, a non-muscle myosin II inhibitor, has been suggested to improve pulmonary endothelial barrier function and control inflammation by suppressing immune cell migration; however, its efficacy in atopic dermatitis is unknown. In this study, we investigated whether (S)-(-)-blebbistatin O-benzoate, a derivative of (-)-blebbistatin, improves dermatitis symptoms in a mite antigen-induced atopic dermatitis model using NC/Nga mice. The efficacy of the compound was confirmed using dermatitis scores, ear thickness measurements, serum IgE levels, histological analysis of lesions, and filaggrin expression analysis, which is important for barrier function. (S)-(-)-Blebbistatin O-benzoate treatment significantly reduced the dermatitis score and serum IgE levels compared to those in the vehicle group (p < 0.05). Furthermore, the histological analysis revealed enhanced filaggrin production and a decreased number of mast cells (p < 0.05), indicating that (S)-(-)-blebbistatin O-benzoate improved atopic dermatitis symptoms in a pathological model. In vitro analysis using cultured keratinocytes revealed increased expression of filaggrin, loricrin, involucrin, and ceramide production pathway-related genes, suggesting that (S)-(-)-blebbistatin O-benzoate promotes epidermal barrier formation. Furthermore, the effect of (S)-(-)-blebbistatin O-benzoate on type 2 alarmin cytokines, which are secreted from epidermal cells upon scratching or allergen stimulation and are involved in the pathogenesis of atopic dermatitis, was evaluated using antigens derived from mite feces. The results showed that (S)-(-)-blebbistatin O-benzoate inhibited the upregulation of these cytokines. Based on the above, (S)-(-)-blebbistatin O-benzoate has the potential to be developed as an atopic dermatitis treatment option that controls dermatitis symptoms by suppressing inflammation and improving barrier function by acting on multiple aspects of the pathogenesis of atopic dermatitis.


Sujet(s)
Benzoates , Cytokines , Eczéma atopique , Épiderme , Protéines filaggrine , Composés hétérocycliques avec 4 noyaux ou plus , Animaux , Humains , Mâle , Souris , Antigènes de Dermatophagoides/immunologie , Benzoates/pharmacologie , Benzoates/usage thérapeutique , Cytokines/métabolisme , Eczéma atopique/traitement médicamenteux , Eczéma atopique/anatomopathologie , Eczéma atopique/métabolisme , Modèles animaux de maladie humaine , Épiderme/effets des médicaments et des substances chimiques , Épiderme/métabolisme , Épiderme/anatomopathologie , Protéines filaggrine/effets des médicaments et des substances chimiques , Composés hétérocycliques avec 4 noyaux ou plus/pharmacologie , Composés hétérocycliques avec 4 noyaux ou plus/usage thérapeutique , Immunoglobuline E/sang , Protéines de filaments intermédiaires/métabolisme , Protéines de filaments intermédiaires/génétique , Kératinocytes/effets des médicaments et des substances chimiques , Kératinocytes/métabolisme , Alarmines/effets des médicaments et des substances chimiques
8.
Curr Med Sci ; 44(3): 475-484, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38748372

RÉSUMÉ

Proton-activated G protein-coupled receptors (GPCRs), initially discovered by Ludwig in 2003, are widely distributed in various tissues. These receptors have been found to modulate the immune system in several inflammatory diseases, including inflammatory bowel disease, atopic dermatitis, and asthma. Proton-activated GPCRs belong to the G protein-coupled receptor family and can detect alternations in extracellular pH. This detection triggers downstream signaling pathways within the cells, ultimately influencing the function of immune cells. In this review, we specifically focused on investigating the immune response of proton-activated GPCRs under inflammatory conditions.


Sujet(s)
Immunomodulation , Inflammation , Récepteurs couplés aux protéines G , Transduction du signal , Récepteurs couplés aux protéines G/métabolisme , Récepteurs couplés aux protéines G/génétique , Récepteurs couplés aux protéines G/immunologie , Humains , Inflammation/métabolisme , Inflammation/immunologie , Animaux , Protons , Asthme/immunologie , Asthme/métabolisme , Eczéma atopique/immunologie , Eczéma atopique/métabolisme , Maladies inflammatoires intestinales/immunologie , Maladies inflammatoires intestinales/métabolisme , Concentration en ions d'hydrogène
9.
Genomics ; 116(4): 110870, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38821220

RÉSUMÉ

The pathophysiology of atopic dermatitis (AD) is complex. CD4+ T cells play an essential role in the development of lesions in AD. However, the underlying mechanism remains unclear. In the present study, we investigated the differentially expressed genes (DEGs) between adult AD lesioned and non-lesioned skin using two datasets from the Gene Expression Omnibus (GEO) database. 62 DEGs were shown to be related to cytokine response. Compared to non-lesioned skin, lesioned skin showed immune infiltration with increased numbers of activated natural killer (NK) cells and CD4+ T memory cells (p < 0.01). We then identified 13 hub genes with a strong association with CD4+ T cells using weighted correlation network analysis. Single-cell analysis of AD detected a novel CD4+ T subcluster, CD4+ tissue residency memory cells (TRMs), which were verified through immunohistochemistry (IHC) to be increased in the dermal area of AD. The significant relationship between CD4+ TRM and AD was assessed through further analyses. FOXO1 and SBNO2, two of the 13 hub genes, were characteristically expressed in the CD4+ TRM, but down-regulated in IFN-γ/TNF-α-induced HaCaT cells, as shown using quantitative polymerase chain reaction (qPCR). Moreover, SBNO2 expression was associated with increased Th1 infiltration in AD (p < 0.05). In addition, genes filtered using Mendelian randomization were positively correlated with CD4+ TRM and were highly expressed in IFN-γ/TNF-α-induced HaCaT cells, as determined using qPCR and western blotting. Collectively, our results revealed that the newly identified CD4+ TRM may be involved in the pathogenesis of adult AD.


Sujet(s)
Lymphocytes T CD4+ , Eczéma atopique , Analyse sur cellule unique , Eczéma atopique/génétique , Eczéma atopique/métabolisme , Eczéma atopique/immunologie , Eczéma atopique/anatomopathologie , Humains , Lymphocytes T CD4+/métabolisme , Lymphocytes T CD4+/immunologie , Adulte , Cellules T mémoire/métabolisme , Cellules T mémoire/immunologie , Peau/métabolisme , Cellules HaCaT , Mémoire immunologique , Mâle , Facteurs de transcription Forkhead/génétique , Facteurs de transcription Forkhead/métabolisme
10.
Int J Mol Sci ; 25(8)2024 Apr 09.
Article de Anglais | MEDLINE | ID: mdl-38673730

RÉSUMÉ

Atopic dermatitis (AD), a chronic inflammatory skin disease, is exacerbated by obesity, yet the precise linking mechanism remains elusive. This study aimed to elucidate how obesity amplifies AD symptoms. We studied skin samples from three mouse groups: sham control, AD, and high-fat (HF) + AD. The HF + AD mice exhibited more severe AD symptoms than the AD or sham control mice. Skin lipidome analysis revealed noteworthy changes in arachidonic acid (AA) metabolism, including increased expression of pla2g4, a key enzyme in AA generation. Genes for phospholipid transport (Scarb1) and acyltransferase utilizing AA as the acyl donor (Agpat3) were upregulated in HF + AD skin. Associations were observed between AA-containing phospholipids and skin lipids containing AA and its metabolites. Furthermore, imbalanced phospholipid metabolism was identified in the HF + AD mice, marked by excessive activation of the AA and phosphatidic acid (PA)-mediated pathway. This imbalance featured increased expression of Plcb1, Plcg1, and Dgk involved in PA generation, along with a decrease in genes converting PA into diglycerol (DG) and CDP-DG (Lpin1 and cds1). This investigation revealed imbalanced phospholipid metabolism in the skin of HF + AD mice, contributing to the heightened inflammatory response observed in HF + AD, shedding light on potential mechanisms linking obesity to the exacerbation of AD symptoms.


Sujet(s)
Eczéma atopique , Alimentation riche en graisse , Modèles animaux de maladie humaine , Obésité , Animaux , Eczéma atopique/métabolisme , Eczéma atopique/étiologie , Eczéma atopique/génétique , Eczéma atopique/anatomopathologie , Obésité/métabolisme , Obésité/génétique , Obésité/complications , Souris , Alimentation riche en graisse/effets indésirables , Peau/métabolisme , Peau/anatomopathologie , Métabolisme lipidique/génétique , Souris de lignée C57BL , Acide arachidonique/métabolisme , Lipidomique/méthodes , Mâle , Phospholipides/métabolisme
11.
Allergy ; 79(6): 1573-1583, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38641894

RÉSUMÉ

BACKGROUND: The antimicrobial ribonuclease RNase 7 is abundantly expressed in the epidermis of lesional skin of atopic dermatitis (AD). Host RNase inhibitor (RI) binds to RNase 7 and blocks its ribonuclease activity. This study aimed to evaluate the impact of RNase 7-RI interactions on AD. METHODS: Cultured human primary keratinocytes, with siRNA-mediated downregulation of RNase 7 and RI, were stimulated with the synthetic RNA polyinosinic-polycytidylic acid (poly I:C). Induction of proinflammatory mediators was analyzed by real-time PCR and ELISA. RI expression in AD non-lesional and lesional skin biopsies and healthy controls was analyzed by real-time PCR and immunostaining. RI protein release in vivo on the AD skin surface was determined by western blot. Antimicrobial and ribonuclease assays were used to investigate the functional role of RI. RESULTS: RNase 7 inhibited the RNA-induced expression of proinflammatory mediators in keratinocytes. Accordingly, downregulation of RNase 7 in keratinocytes enhanced RNA-mediated induction of proinflammatory mediators, whereas downregulation of RI had the opposite effect. RI was released by damaged keratinocytes and epidermis. In vivo expression and release of RI on the skin surface were enhanced in lesional AD skin. Rinsing solution from the surface of lesional AD skin blocked the ribonuclease activity of RNase 7. The anti-Staphylococcus aureus activity of RNase 7 was abrogated by RI. CONCLUSIONS: Our data suggest a novel role of RI as a trigger factor of inflammation in AD by blocking the ribonuclease and antimicrobial activity of RNase 7, thereby enhancing RNA-mediated inflammation and S. aureus growth.


Sujet(s)
Eczéma atopique , Kératinocytes , Ribonucléases , Staphylococcus aureus , Humains , Eczéma atopique/métabolisme , Ribonucléases/métabolisme , Kératinocytes/métabolisme , Inflammation/métabolisme , Cellules cultivées
12.
Cells ; 13(7)2024 Mar 28.
Article de Anglais | MEDLINE | ID: mdl-38607026

RÉSUMÉ

The transmembrane glycoprotein OX40 receptor (OX40) and its ligand, OX40L, are instrumental modulators of the adaptive immune response in humans. OX40 functions as a costimulatory molecule that promotes T cell activation, differentiation, and survival through ligation with OX40L. T cells play an integral role in the pathogenesis of several inflammatory skin conditions, including atopic dermatitis (AD). In particular, T helper 2 (TH2) cells strongly contribute to AD pathogenesis via the production of cytokines associated with type 2 inflammation (e.g., IL-4, IL-5, IL-13, and IL-31) that lead to skin barrier dysfunction and pruritus. The OX40-OX40L interaction also promotes the activation and proliferation of other T helper cell populations (e.g., TH1, TH22, and TH17), and AD patients have demonstrated higher levels of OX40 expression on peripheral blood mononuclear cells than healthy controls. As such, the OX40-OX40L pathway is a potential target for AD treatment. Novel therapies targeting the OX40 pathway are currently in development, several of which have demonstrated promising safety and efficacy results in patients with moderate-to-severe AD. Herein, we review the function of OX40 and the OX40-OX40L signaling pathway, their role in AD pathogenesis, and emerging therapies targeting OX40-OX40L that may offer insights into the future of AD management.


Sujet(s)
Eczéma atopique , Humains , Différenciation cellulaire , Cytokines/métabolisme , Eczéma atopique/métabolisme , Eczéma atopique/anatomopathologie , Inflammation , Agranulocytes/métabolisme
13.
Exp Dermatol ; 33(4): e15079, 2024 Apr.
Article de Anglais | MEDLINE | ID: mdl-38654506

RÉSUMÉ

Common characteristics in the pathogenesis of psoriasis (PS) and atopic dermatitis (AD) have been presumed, but only a few studies have clearly supported this. The current aim was to find possible similarities and differences in protein expression patterns between these two major chronic inflammatory skin diseases. High-throughput tandem mass spectrometry proteomic analysis was performed using full thickness skin samples from adult PS patients, AD patients and healthy subjects. We detected a combined total of 3045 proteins in the three study groups. According to principal component analysis, there was significant overlap between the proteomic profiles of PS and AD, and both clearly differed from that of healthy skin. The following validation of selected proteins with western blot analysis showed similar tendencies in expression levels and produced statistically significant results. The expression of periostin (POSTN) was consistently high in AD and very low or undetectable in PS (5% FDR corrected p < 0.001), suggesting POSTN as a potential biomarker to distinguish these diseases. Immunohistochemistry further confirmed higher POSTN expression in AD compared to PS skin. Overall, our findings support the concept that these two chronic skin diseases might share considerably more common mechanisms in pathogenesis than has been suspected thus far.


Sujet(s)
Molécules d'adhérence cellulaire , Eczéma atopique , Protéomique , Psoriasis , Eczéma atopique/métabolisme , Humains , Psoriasis/métabolisme , Protéomique/méthodes , Molécules d'adhérence cellulaire/métabolisme , Adulte , Femelle , Mâle , Adulte d'âge moyen , Marqueurs biologiques/métabolisme , Spectrométrie de masse en tandem , Peau/métabolisme , Analyse en composantes principales , Études cas-témoins
15.
J Allergy Clin Immunol ; 153(5): 1456-1463.e4, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38442771

RÉSUMÉ

BACKGROUND: Food allergy (FA) often occurs in early childhood with and without atopic dermatitis (AD). FA can be severe and even fatal. For primary prevention, it is important to find early biomarkers to predict the future onset of FA before any clinical manifestations. OBJECTIVE: Our aim was to find early predictors of future onset of FA in the stratum corneum (SC). METHODS: Skin tape strips were collected from the forearm of newborns (n = 129) at age 2 months, before any signs of clinical FA or AD. Children were clinically monitored until they reached age 2 years to confirm the presence or absence of FA and AD. Skin tape strips were subjected to lipidomic analyses by liquid chromatography-tandem mass spectrometry and cytokine determination by Meso Scale Discovery U-Plex assay. RESULTS: Overall, 9 of 129 infants (7.0%) developed FA alone and 9 of 129 infants (7.0%) developed FA concomitantly with AD. In the stratum corneum of children with future FA and concomitant AD and FA, absolute amounts of unsaturated (N24:1)(C18-sphingosine)ceramide and (N26:1)(C18-sphingosine)ceramide and their relative percentages within the molecular group were increased compared with the amounts and percentages in healthy children, with P values ranging from less than .01 to less than .05 according to ANOVA. The children with future AD had normal levels of these molecules. IL-33 level was upregulated in those infants with future FA but not in those with future AD, whereas thymic stromal lymphopoietin was upregulated in those with future AD but not in those with future FA. Logistic regression analysis revealed strong FA predicting power for the combination of dysregulated lipids and cytokines, with an odds ratio reaching 101.4 (95% CI = 5.4-1910.6). CONCLUSION: Noninvasive skin tape strip analysis at age 2 months can identify infants at risk of FA in the future.


Sujet(s)
Marqueurs biologiques , Cytokines , Eczéma atopique , Hypersensibilité alimentaire , Humains , Nourrisson , Hypersensibilité alimentaire/immunologie , Hypersensibilité alimentaire/diagnostic , Mâle , Femelle , Eczéma atopique/immunologie , Eczéma atopique/métabolisme , Cytokines/métabolisme , Nouveau-né , Peau/immunologie , Peau/métabolisme , Enfant d'âge préscolaire , Céramides/métabolisme , Céramides/analyse
16.
Crit Rev Eukaryot Gene Expr ; 34(4): 1-11, 2024.
Article de Anglais | MEDLINE | ID: mdl-38505868

RÉSUMÉ

Keratin 6A (KRT6A) is involved in the pathogenesis of various skin diseases. However, the reports on the roles of KRT6A in atopic dermatitis (AD) are limited. This study aimed to investigate the potentials of KRT6A in AD. mRNA levels were detected by RT-PCR. Cytokine release was determined by ELISA. Protein expression was determined using Western blot. Cell viability was determined by CCK-8. Cytotoxicity was detected by LDH assay. Cell death was determined by TUNEL. The pyroptosis of keratinocytes was detected using flow cytometry. We found that KRT6A was overexpressed in AD patients. Moreover, KRT6A was stimulated after exposed to proinflammatory cytokines. Overexpressed KRT6A suppressed inflammatory response, while KRT6A knockdown exerted the opposite effects. Overexpressed KRT6A suppressed inflammation-induced pyroptosis of keratinocytes. Additionally, KRT6A negatively regulated interleukin-17a (IL-17a) expression, blocking IL-17 signaling. IL-17a overexpression antagonized the effects of KRT6A and promoted pyroptosis of keratinocytes. In conclusion, KRT6A exerted protective functions in AD via regulating IL-17 signaling. This KRT6A/IL-17 may be a novel target for AD.


Sujet(s)
Eczéma atopique , Interleukine-17 , Humains , Interleukine-17/génétique , Interleukine-17/métabolisme , Interleukine-17/pharmacologie , Pyroptose , Kératine-6/métabolisme , Kératine-6/pharmacologie , Kératinocytes/métabolisme , Transduction du signal , Cytokines/métabolisme , Eczéma atopique/génétique , Eczéma atopique/métabolisme
17.
Xenobiotica ; 54(3): 138-149, 2024 Mar.
Article de Anglais | MEDLINE | ID: mdl-38501457

RÉSUMÉ

HY-072808 is a novel phosphodiesterase 4 inhibitor clinically used for topical atopic dermatitis treatment. Cytochrome P450 enzymes are involved in transforming it into major metabolite ZZ-24. An efficient UPLC-MS/MS method was established to detect HY-072808 and ZZ-24 in plasma and skin tissues of minipigs.One-step protein precipitation was performed with acetonitrile. Subsequently, elution was served with a methanol and water gradient containing 0.1% formic acid for 3.5 min. The plasma and skin tissue concentrations of HY-072808 and ZZ-24 showed good linearity from 0.200 to 200 ng/mL.The experimental minipigs exhibited low systemic exposure and bioavailability of 3.1-7.6% after transdermal application of 1-4% HY-072808 ointment. Multiple topical administrations over seven consecutive days showed a minor accumulation in systemic exposure, with accumulation factors of 2.3 and 4.0 for HY-072808 and ZZ-24, respectively.The distribution of HY-072808 ointment among different cortical layers in minipigs was studied for the first time. Following transdermal application of 2% HY-072808 ointment, the concentration in plasma and skin tissues in the order of epidermis > dermis > subcutaneous tissue ≈ subcutaneous muscle ≈ plasma; at 48 h after the administration, the epidermis and dermis still had a high concentration of the drug.


Sujet(s)
Eczéma atopique , Animaux , Suidae , Porc miniature/métabolisme , Préparations pharmaceutiques/métabolisme , Eczéma atopique/traitement médicamenteux , Eczéma atopique/métabolisme , Chromatographie en phase liquide , Biodisponibilité , , Onguents/usage thérapeutique , Spectrométrie de masse en tandem/méthodes
18.
Sci Rep ; 14(1): 6263, 2024 03 15.
Article de Anglais | MEDLINE | ID: mdl-38491103

RÉSUMÉ

Psychological stress and intestinal leakage are key factors in atopic dermatitis (AD) recurrence and exacerbation. Here, we demonstrate the mechanism underlying bacterial translocation across intestinal epithelial barrier damaged due to stress and further aggravation of trimellitic anhydride (TMA)-induced itch, which remain unclear, in AD mice. Immobilization (IMO) stress exacerbated scratching bouts and colon histological damage, and increased serum corticosterone and lipopolysaccharide (LPS). Orally administered fluorescein isothiocyanate (FITC)-dextran and surgically injected (into the colon) Cy5.5-conjugated LPS were detected in the serum and skin after IMO stress, respectively. The relative abundance of aerobic or facultative anaerobic bacteria was increased in the colon mucus layer, and Lactobacillus murinus, E. coli, Staphylococcus nepalensis, and several strains of Bacillus sp. were isolated from the spleens and mesenteric lymph nodes. Oral antibiotics or intestinal permeability blockers, such as lubiprostone (Lu), 2,4,6-triaminopyrimidine (TAP) and ML-7, inhibited IMO stress-associated itch; however, it was reinduced through intradermal or i.p. injection of LPS without IMO stress. I.p. injection of TAK-242 (resatorvid), a TLR4 inhibitor, abrogated IMO stress-associated itch, which was also confirmed in TLR4-KO mice. IMO stress alone did not cause itch in naïve mice. IMO stress-induced itch aggravation in TMA-treated AD mice might be attributed to the translocation of gut-derived bacterial cells and LPS, which activates peripheral TLR4 signaling.


Sujet(s)
Eczéma atopique , Récepteur de type Toll-4 , Animaux , Souris , Eczéma atopique/métabolisme , Eczéma atopique/anatomopathologie , Modèles animaux de maladie humaine , Escherichia coli , Lipopolysaccharides/métabolisme , Prurit/induit chimiquement , Récepteur de type Toll-4/métabolisme
19.
Nat Commun ; 15(1): 1666, 2024 Feb 23.
Article de Anglais | MEDLINE | ID: mdl-38396021

RÉSUMÉ

Both monocytes and macrophages are heterogeneous populations. It was traditionally understood that Ly6Chi classical (inflammatory) monocytes differentiate into pro-inflammatory Ly6Chi macrophages. Accumulating evidence has suggested that Ly6Chi classical monocytes can also differentiate into Ly6Clo pro-resolving macrophages under certain conditions, while their differentiation trajectory remains to be fully elucidated. The present study with scRNA-seq and flow cytometric analyses reveals that Ly6ChiPD-L2lo classical monocytes recruited to the allergic skin lesion sequentially differentiate into Ly6CloPD-L2hi pro-resolving macrophages, via intermediate Ly6ChiPD-L2hi macrophages but not Ly6Clo non-classical monocytes, in an IL-4 receptor-dependent manner. Along the differentiation, classical monocyte-derived macrophages display anti-inflammatory signatures followed by metabolic rewiring concordant with their ability to phagocytose apoptotic neutrophils and allergens, therefore contributing to the resolution of inflammation. The failure in the generation of these pro-resolving macrophages drives the IL-1α-mediated cycle of inflammation with abscess-like accumulation of necrotic neutrophils. Thus, we clarify the stepwise differentiation trajectory from Ly6Chi classical monocytes toward Ly6Clo pro-resolving macrophages that restrain neutrophilic aggravation of skin allergic inflammation.


Sujet(s)
Eczéma atopique , Monocytes , Souris , Animaux , Monocytes/métabolisme , Macrophages/métabolisme , Inflammation/anatomopathologie , Analyse de profil d'expression de gènes , Eczéma atopique/métabolisme , Souris de lignée C57BL
20.
Cells ; 13(3)2024 Jan 25.
Article de Anglais | MEDLINE | ID: mdl-38334612

RÉSUMÉ

Mast cells (MCs) are an important part of the immune system, responding both to pathogens and toxins, but they also play an important role in allergic diseases, where recent data show that non-IgE-mediated activation is also of relevance, especially in chronic urticaria (CU) and atopic dermatitis (AD). Skin MCs express Mas-related G-protein-coupled receptor X2 (MRGPRX2), a key protein in non-IgE-dependent MC degranulation, and its overactivity is one of the triggering factors for the above-mentioned diseases, making MRGPRX2 a potential therapeutic target. Reviewing the latest literature revealed our need to focus on the discovery of MRGPRX2 activators as well as the ongoing vast research towards finding specific MRGPRX2 inhibitors for potential therapeutic approaches. Most of these studies are in their preliminary stages, with one drug currently being investigated in a clinical trial. Future studies and improved model systems are needed to verify whether any of these inhibitors may have the potential to be the next therapeutic treatment for CU, AD, and other pseudo-allergic reactions.


Sujet(s)
Urticaire chronique , Eczéma atopique , Hypersensibilité , Humains , Eczéma atopique/traitement médicamenteux , Eczéma atopique/métabolisme , Hypersensibilité/métabolisme , Mastocytes/métabolisme , Urticaire chronique/traitement médicamenteux , Récepteurs couplés aux protéines G/métabolisme , Protéines de tissu nerveux/métabolisme , Récepteur aux neuropeptides/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...