Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 1.180
Filtrer
1.
Front Immunol ; 15: 1393799, 2024.
Article de Anglais | MEDLINE | ID: mdl-38975347

RÉSUMÉ

SOCS are a family of negative inhibitors of the molecular cascades induced by cytokines, growth factors and hormones. At molecular level, SOCS proteins inhibit the kinase activity of specific sets of receptor-associated Janus Activated Kinases (JAKs), thereby suppressing the propagation of intracellular signals. Of the eight known members, SOCS1 and SOCS3 inhibit activity of JAKs mainly induced by cytokines and can play key roles in regulation of inflammatory and immune responses. SOCS1 and SOCS3 are the most well-characterized SOCS members in skin inflammatory diseases, where their inhibitory activity on cytokine activated JAKs and consequent anti-inflammatory action has been widely investigated in epidermal keratinocytes. Structurally, SOCS1 and SOCS3 share the presence of a N-terminal domain containing a kinase inhibitory region (KIR) motif able to act as a pseudo-substrate for JAK and to inhibit its activity. During the last decades, the design and employment of SOCS1 and SOCS3-derived peptides mimicking KIR domains in experimental models of dermatoses definitively established a strong anti-inflammatory and ameliorative impact of JAK inhibition on skin inflammatory responses. Herein, we discuss the importance of the findings collected in the past on SOCS1 and SOCS3 function in the inflammatory responses associated to skin immune-mediated diseases and malignancies, for the development of the JAK inhibitor drugs. Among them, different JAK inhibitors have been introduced in the clinical practice for treatment of atopic dermatitis and psoriasis, and others are being investigated for skin diseases like alopecia areata and vitiligo.


Sujet(s)
Transformation cellulaire néoplasique , Protéine-1 suppressive de la signalisation des cytokines , Protéine-3 suppressive de la signalisation des cytokine , Humains , Protéine-1 suppressive de la signalisation des cytokines/métabolisme , Animaux , Protéine-3 suppressive de la signalisation des cytokine/métabolisme , Transformation cellulaire néoplasique/immunologie , Transformation cellulaire néoplasique/métabolisme , Transduction du signal , Tumeurs cutanées/immunologie , Tumeurs cutanées/métabolisme , Tumeurs cutanées/anatomopathologie , Dermatite/immunologie , Dermatite/métabolisme , Janus kinases/métabolisme , Peau/immunologie , Peau/anatomopathologie , Peau/métabolisme
2.
Arch Dermatol Res ; 316(7): 348, 2024 Jun 07.
Article de Anglais | MEDLINE | ID: mdl-38849562

RÉSUMÉ

This study investigates the mechanism through which paeoniflorin inhibits TSLP expression to regulate dendritic cell activation in corticosteroid-dependent dermatitis treatment. Utilizing databases like TCMSP, we identified paeoniflorin's components, targets, and constructed networks. Molecular docking and gene enrichment analysis helped pinpoint key targets and pathways affected by paeoniflorin. In vitro and in vivo models were used to study CD80, CD86, cytokines, T-cell activation, skin lesions, histopathological changes, TSLP, CD80, and CD86 expression. Our study revealed paeoniflorin's active constituent targeting IL-6 in corticosteroid-dependent dermatitis. In vitro experiments demonstrated reduced TSLP expression, CD80, CD86, and cytokine secretion post-paeoniflorin treatment. In vivo, paeoniflorin significantly decreased skin lesion severity, cytokine levels, TSLP, CD80, and CD86 expression. The study highlights paeoniflorin's efficacy in inhibiting TSLP expression and suppressing dendritic cell activation in corticosteroid-dependent dermatitis, suggesting its potential as a therapeutic intervention. Additionally, it offers insights into the complex molecular mechanisms underlying paeoniflorin's anti-inflammatory properties in treating corticosteroid-dependent dermatitis.


Sujet(s)
Cytokines , Cellules dendritiques , Glucosides , Monoterpènes , Cellules dendritiques/effets des médicaments et des substances chimiques , Cellules dendritiques/immunologie , Cellules dendritiques/métabolisme , Glucosides/pharmacologie , Glucosides/usage thérapeutique , Animaux , Cytokines/métabolisme , Monoterpènes/pharmacologie , Monoterpènes/usage thérapeutique , Humains , Souris , Dermatite/traitement médicamenteux , Dermatite/immunologie , Dermatite/métabolisme , Interleukine-6/métabolisme , Simulation de docking moléculaire , Peau/anatomopathologie , Peau/effets des médicaments et des substances chimiques , Peau/immunologie , Peau/métabolisme , Anti-inflammatoires/pharmacologie , Anti-inflammatoires/usage thérapeutique , Modèles animaux de maladie humaine , Antigène CD80/métabolisme , Antigène CD86/métabolisme , Mâle , Lymphopoïétine stromale thymique , Activation des lymphocytes/effets des médicaments et des substances chimiques
3.
J Exp Med ; 221(8)2024 Aug 05.
Article de Anglais | MEDLINE | ID: mdl-38861030

RÉSUMÉ

Germline gain-of-function (GOF) variants in STAT3 cause an inborn error of immunity associated with early-onset poly-autoimmunity and immune dysregulation. To study tissue-specific immune dysregulation, we used a mouse model carrying a missense variant (p.G421R) that causes human disease. We observed spontaneous and imiquimod (IMQ)-induced skin inflammation associated with cell-intrinsic local Th17 responses in STAT3 GOF mice. CD4+ T cells were sufficient to drive skin inflammation and showed increased Il22 expression in expanded clones. Certain aspects of disease, including increased epidermal thickness, also required the presence of STAT3 GOF in epithelial cells. Treatment with a JAK inhibitor improved skin disease without affecting local Th17 recruitment and cytokine production. These findings collectively support the involvement of Th17 responses in the development of organ-specific immune dysregulation in STAT3 GOF and suggest that the presence of STAT3 GOF in tissues is important for disease and can be targeted with JAK inhibition.


Sujet(s)
Mutation gain de fonction , Imiquimod , Facteur de transcription STAT-3 , Cellules Th17 , Animaux , Facteur de transcription STAT-3/métabolisme , Facteur de transcription STAT-3/génétique , Cellules Th17/immunologie , Souris , Humains , Imiquimod/pharmacologie , Peau/anatomopathologie , Peau/métabolisme , Peau/immunologie , , Dermatite/immunologie , Dermatite/génétique , Dermatite/anatomopathologie , Dermatite/métabolisme , Souris de lignée C57BL , Interleukines/génétique , Interleukines/métabolisme , Lymphocytes T CD4+/immunologie , Lymphocytes T CD4+/métabolisme , Inflammation/génétique , Inflammation/métabolisme , Inflammation/immunologie , Inflammation/anatomopathologie
4.
Pharmacol Res ; 205: 107231, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38815878

RÉSUMÉ

We previously demonstrated that mice carrying natural mtDNA variants of the FVB/NJ strain (m.7778 G>T in the mt-Atp8 gene in mitochondrial complex V), namely C57BL/6 J-mtFVB/NJ (B6-mtFVB), exhibited (i) partial protection from experimental skin inflammatory diseases in an anti-murine type VII collagen antibody-induced skin inflammation model and psoriasiform dermatitis model; (ii) significantly altered metabolites, including short-chain fatty acids, according to targeted metabolomics of liver, skin and lymph node samples; and (iii) a differential composition of the gut microbiota according to bacterial 16 S rRNA gene sequencing of stool samples compared to wild-type C57BL/6 J (B6) mice. To further dissect these disease-contributing factors, we induced an experimental antibody-induced skin inflammatory disease in gnotobiotic mice. We performed shotgun metagenomic sequencing of caecum contents and untargeted metabolomics of liver, CD4+ T cell, and caecum content samples from conventional B6-mtFVB and B6 mice. We identified D-glucosamine as a candidate mediator that ameliorated disease severity in experimental antibody-induced skin inflammation by modulating immune cell function in T cells, neutrophils and macrophages. Because mice carrying mtDNA variants of the FVB/NJ strain show differential disease susceptibility to a wide range of experimental diseases, including diet-induced atherosclerosis in low-density lipoprotein receptor knockout mice and collagen antibody-induced arthritis in DBA/1 J mice, this experimental approach is valuable for identifying novel therapeutic options for skin inflammatory conditions and other chronic inflammatory diseases to which mice carrying specific mtDNA variants show differential susceptibility.


Sujet(s)
ADN mitochondrial , Souris de lignée C57BL , Animaux , ADN mitochondrial/génétique , Microbiome gastro-intestinal , Souris , Peau/métabolisme , Peau/microbiologie , Peau/anatomopathologie , Dermatite/immunologie , Dermatite/microbiologie , Dermatite/génétique , Dermatite/traitement médicamenteux , Dermatite/métabolisme , Inflammation/génétique , Inflammation/immunologie , Modèles animaux de maladie humaine , Mâle , Axénie , Psoriasis/traitement médicamenteux , Psoriasis/immunologie , Psoriasis/génétique , Caecum/microbiologie , Maladie chronique , Femelle
5.
Int Immunopharmacol ; 134: 112261, 2024 Jun 15.
Article de Anglais | MEDLINE | ID: mdl-38761783

RÉSUMÉ

BACKGROUND: Psoriasis, a chronic inflammatory condition of the skin, is characterized by an atypical proliferation of epidermal keratinocytes and immune cell infiltration. Orientin is a flavonoid monomer with potent anti-inflammatory activities. However, the therapeutic effects of orientin on psoriasis and the underlying mechanisms have not been elucidated. OBJECTIVE: To investigate the therapeutic effect of orientin on psoriasis and the underlying mechanisms using network pharmacology and experimental studies. METHODS: A psoriasis-like mouse model was established using imiquimod (IMQ). Lipopolysaccharide (LPS) was used to stimulate the RAW264.7 and HaCaT cells in vitro. The therapeutic effects of orientin and the underlying mechanism were analyzed using histopathological, immunohistochemical, quantitative real-time polymerase chain reaction, enzyme-linked immunosorbent assay, flow cytometry, and western blotting analyses. RESULTS: Orientin ameliorated skin lesions and suppressed keratinocyte proliferation and immune cell infiltration in the IMQ-induced psoriasis-like mouse model. Additionally, orientin inhibited the secretion of the pro-inflammatory factors interleukin (IL)-1ß, tumor necrosis factor (TNF)-α, IL-6, IL-8, IL-17, and IL-23 in the psoriasis-like mouse model and LPS-induced RAW264.7 and HaCaT cells. Furthermore, orientin mitigated the LPS-induced upregulation of reactive oxygen species and downregulation of IL-10 and glutathione levels. Orientin alleviated inflammation by downregulating the MAPK signaling pathway. CONCLUSION: Orientin alleviated psoriasis-like dermatitis by suppressing the MAPK signaling pathway, suggesting that orientin is a potential therapeutic for psoriasis.


Sujet(s)
Anti-inflammatoires , Cytokines , Modèles animaux de maladie humaine , Flavonoïdes , Glucosides , Cellules HaCaT , Imiquimod , Kératinocytes , Lipopolysaccharides , Système de signalisation des MAP kinases , Souris de lignée BALB C , Psoriasis , Animaux , Psoriasis/traitement médicamenteux , Psoriasis/immunologie , Psoriasis/induit chimiquement , Psoriasis/anatomopathologie , Souris , Humains , Cellules RAW 264.7 , Anti-inflammatoires/usage thérapeutique , Anti-inflammatoires/pharmacologie , Flavonoïdes/pharmacologie , Flavonoïdes/usage thérapeutique , Cytokines/métabolisme , Kératinocytes/effets des médicaments et des substances chimiques , Glucosides/usage thérapeutique , Glucosides/pharmacologie , Système de signalisation des MAP kinases/effets des médicaments et des substances chimiques , Peau/anatomopathologie , Peau/effets des médicaments et des substances chimiques , Peau/immunologie , Prolifération cellulaire/effets des médicaments et des substances chimiques , Mâle , Espèces réactives de l'oxygène/métabolisme , Dermatite/traitement médicamenteux , Dermatite/anatomopathologie , Dermatite/immunologie , Lignée cellulaire
7.
PLoS One ; 19(4): e0302555, 2024.
Article de Anglais | MEDLINE | ID: mdl-38683795

RÉSUMÉ

Clostridial dermatitis (CD), caused by Clostridium septicum, is an emerging disease of increasing economic importance in turkeys. Currently, there are no effective vaccines for CD control. Here, two non-toxic domains of C. septicum alpha toxin, namely ntATX-D1 and ntATX-D2, were identified, cloned, and expressed in Escherichia coli as recombinant subunit proteins to investigate their use as potential vaccine candidates. Experimental groups consisted of a Negative control (NCx) that did not receive C. septicum challenge, while the adjuvant-only Positive control (PCx), ntATX-D1 immunization (D1) and ntATX-D2 immunization (D2) groups received C. septicum challenge. Turkeys were immunized subcutaneously with 100 µg of protein at 7, 8 and 9 weeks of age along with an oil-in-water nano-emulsion adjuvant, followed by C. septicum challenge at 11 weeks of age. Results showed that while 46.2% of birds in the PCx group died post-challenge, the rate of mortality in D1- or D2-immunization groups was 13.3%. The gross and histopathological lesions in the skin, muscle and spleen showed that the disease severity was highest in PCx group, while the D2-immunized birds had significantly lower lesion scores when compared to PCx. Gene expression analysis revealed that PCx birds had significantly higher expression of pro-inflammatory cytokine genes in the skin, muscle and spleen than the NCx group, while the D2 group had significantly lower expression of these genes compared to PCx. Peripheral blood cellular analysis showed increased frequencies of activated CD4+ and/or CD8+ cells in the D1 and D2-immunized groups. Additionally, the immunized turkeys developed antigen-specific serum IgY antibodies. Collectively, these findings indicate that ntATX proteins, specifically the ntATX-D2 can be a promising vaccine candidate for protecting turkeys against CD and that the protection mechanisms may include downregulation of C. septicum-induced inflammation and increased CD4+ and CD8+ cellular activation.


Sujet(s)
Toxines bactériennes , Infections à Clostridium , Clostridium septicum , Dermatite , Maladies de la volaille , Protéines recombinantes , Dindons , Animaux , Dindons/immunologie , Clostridium septicum/immunologie , Infections à Clostridium/prévention et contrôle , Infections à Clostridium/immunologie , Infections à Clostridium/médecine vétérinaire , Maladies de la volaille/prévention et contrôle , Maladies de la volaille/immunologie , Maladies de la volaille/microbiologie , Toxines bactériennes/immunologie , Protéines recombinantes/immunologie , Protéines recombinantes/administration et posologie , Dermatite/prévention et contrôle , Dermatite/immunologie , Dermatite/médecine vétérinaire , Vaccins antibactériens/immunologie , Vaccins antibactériens/administration et posologie , Immunisation
8.
JCI Insight ; 9(8)2024 Mar 12.
Article de Anglais | MEDLINE | ID: mdl-38470486

RÉSUMÉ

IL-17C is an epithelial cell-derived proinflammatory cytokine whose transcriptional regulation remains unclear. Analysis of the IL17C promoter region identified TCF4 as putative regulator, and siRNA knockdown of TCF4 in human keratinocytes (KCs) increased IL17C. IL-17C stimulation of KCs (along with IL-17A and TNF-α stimulation) decreased TCF4 and increased NFKBIZ and ZC3H12A expression in an IL-17RA/RE-dependent manner, thus creating a feedback loop. ZC3H12A (MCPIP1/Regnase-1), a transcriptional immune-response regulator, also increased following TCF4 siRNA knockdown, and siRNA knockdown of ZC3H12A decreased NFKBIZ, IL1B, IL36G, CCL20, and CXCL1, revealing a proinflammatory role for ZC3H12A. Examination of lesional skin from the KC-Tie2 inflammatory dermatitis mouse model identified decreases in TCF4 protein concomitant with increases in IL-17C and Zc3h12a that reversed following the genetic elimination of Il17c, Il17ra, and Il17re and improvement in the skin phenotype. Conversely, interference with Tcf4 in KC-Tie2 mouse skin increased Il17c and exacerbated the inflammatory skin phenotype. Together, these findings identify a role for TCF4 in the negative regulation of IL-17C, which, alone and with TNF-α and IL-17A, feed back to decrease TCF4 in an IL-17RA/RE-dependent manner. This loop is further amplified by IL-17C-TCF4 autocrine regulation of ZC3H12A and IL-17C regulation of NFKBIZ to promote self-sustaining skin inflammation.


Sujet(s)
Protéines adaptatrices de la transduction du signal , Interleukine-17 , Kératinocytes , Récepteurs à l'interleukine-17 , Ribonucléases , Transduction du signal , Facteur-4 de transcription , Animaux , Facteur-4 de transcription/métabolisme , Facteur-4 de transcription/génétique , Humains , Interleukine-17/métabolisme , Interleukine-17/génétique , Souris , Kératinocytes/métabolisme , Ribonucléases/métabolisme , Ribonucléases/génétique , Récepteurs à l'interleukine-17/métabolisme , Récepteurs à l'interleukine-17/génétique , Inflammation/métabolisme , Inflammation/génétique , Modèles animaux de maladie humaine , Épiderme/métabolisme , Dermatite/métabolisme , Dermatite/génétique , Dermatite/immunologie , Dermatite/anatomopathologie , Rétrocontrôle physiologique , Régulation de l'expression des gènes
10.
Dermatology ; 239(2): 248-254, 2023.
Article de Anglais | MEDLINE | ID: mdl-36652928

RÉSUMÉ

BACKGROUND: Adult-onset immunodeficiency (AOID) due to interferon-gamma autoantibody is a rare, acquired immunodeficiency disease. Reactive neutrophilic dermatoses (RND), predominantly Sweet syndrome (SS), and generalized pustular eruption have been reported repeatedly. OBJECTIVES: The aims of this study were to describe the cutaneous manifestations in AOID patients and determine the incidence of RND and associated factors using a larger population size than have been previously reported. METHODS: A retrospective chart review of all confirmed AOID cases in Chiang Mai University Hospital from January 2006 to June 2020 was conducted. The demographics and characteristics of RND including type, onset, and laboratory information in every episode of cutaneous manifestations were collected. Generalized estimating equations of binary logistic regression were used to determine the indicators of RND. RESULTS: A total of 146 patients with confirmed AOID were identified. Of these, 57 cases (39%) developed at least one episode of RND. Thirteen cases (23%) of the patients experienced RND twice during the follow-up period. All recurrence of RND displayed the same cutaneous phenotype, with the exception of 2 cases who had both SS and generalized pustular eruption. Finally, 49 episodes of SS and 22 episodes of generalized pustular eruption were included in the analysis. All patients with RND had concomitant active opportunistic infections, of which most were non-tuberculous mycobacterium (NTM) infection. NTM infection (prevalence odds ratio [POR] 2.87), lymphadenopathy (POR 3.30) as well as lower serum alkaline phosphatase (ALP) level (POR 0.71 for every 100-unit increment in ALP) were found to be significantly associated with RND occurrence. CONCLUSIONS: 39% of our AOID patients experienced RND once during the course of the disease. Notable factors associated with RND occurrence were concomitant NTM infection, lymphadenopathy, and lower level of ALP.


Sujet(s)
Dermatite , Déficits immunitaires , Humains , Autoanticorps , Dermatite/étiologie , Dermatite/immunologie , Déficits immunitaires/complications , Déficits immunitaires/épidémiologie , Interféron gamma/immunologie , Lymphadénopathie/complications , Études rétrospectives , Syndrome de Sweet/étiologie , Syndrome de Sweet/complications , Granulocytes neutrophiles/immunologie , Granulocytes neutrophiles/anatomopathologie
11.
Proc Natl Acad Sci U S A ; 119(26): e2200348119, 2022 06 28.
Article de Anglais | MEDLINE | ID: mdl-35727974

RÉSUMÉ

Immune checkpoint inhibitors (ICIs) are essential components of the cancer therapeutic armamentarium. While ICIs have demonstrated remarkable clinical responses, they can be accompanied by immune-related adverse events (irAEs). These inflammatory side effects are of unclear etiology and impact virtually all organ systems, with the most common being sites colonized by the microbiota such as the skin and gastrointestinal tract. Here, we establish a mouse model of commensal bacteria-driven skin irAEs and demonstrate that immune checkpoint inhibition unleashes commensal-specific inflammatory T cell responses. These aberrant responses were dependent on production of IL-17 by commensal-specific T cells and induced pathology that recapitulated the cutaneous inflammation seen in patients treated with ICIs. Importantly, aberrant T cell responses unleashed by ICIs were sufficient to perpetuate inflammatory memory responses to the microbiota months following the cessation of treatment. Altogether, we have established a mouse model of skin irAEs and reveal that ICIs unleash aberrant immune responses against skin commensals, with long-lasting inflammatory consequences.


Sujet(s)
Dermatite , Inhibiteurs de points de contrôle immunitaires , Microbiote , Animaux , Dermatite/immunologie , Dermatite/microbiologie , Modèles animaux de maladie humaine , Inhibiteurs de points de contrôle immunitaires/effets indésirables , Immunité/effets des médicaments et des substances chimiques , Interleukine-17/métabolisme , Souris , Microbiote/effets des médicaments et des substances chimiques , Microbiote/immunologie , Staphylococcus epidermidis/effets des médicaments et des substances chimiques , Staphylococcus epidermidis/immunologie , Symbiose/effets des médicaments et des substances chimiques , Lymphocytes T/immunologie
12.
J Invest Dermatol ; 142(3 Pt B): 768-773, 2022 03.
Article de Anglais | MEDLINE | ID: mdl-34130802

RÉSUMÉ

Autoimmunity results from the breaking of immune tolerance, leading to inflammation and pathology. Although well studied in the conventional T-cell field, the role of nonconventional T cells in autoimmunity is less understood. CD1-restricted T cells recognize lipid antigens rather than peptide antigens and have been implicated in various autoimmune skin conditions, including psoriasis and atopic dermatitis. In this review, we will discuss the self-lipids that CD1-restricted T cells recognize and how these T cells become aberrantly regulated in pathogenic skin conditions.


Sujet(s)
Antigènes CD1 , Dermatite/immunologie , Lymphocytes T , Présentation d'antigène , Auto-immunité , Tolérance immunitaire
13.
Am J Dermatopathol ; 43(12): e222-e226, 2021 Dec 01.
Article de Anglais | MEDLINE | ID: mdl-34797806

RÉSUMÉ

ABSTRACT: Paraneoplastic granulomatous disease occurs in approximately 7.3% of patients with non-Hodgkin lymphoma, most commonly among patients with chronic lymphocytic leukemia (CLL). These lesions are often reported to appear similar to sarcoidosis in clinical presentation and under light microscopy. However, comprehensive descriptions of the cytomorphologic characteristics of these paraneoplastic granulomas are lacking, and the mechanisms involved in their formation remain ill-defined. Noninfectious dermal granulomatous reactions have also been reported in many primary immunodeficiencies, including common variable immune deficiency and ataxia-telangiectasia. We present a case of noninfectious CD8+ predominant granulomatous dermatitis with ocular involvement occurring in the setting of CLL and marked hypogammaglobulinemia. Based on the analysis of shared factors in patients with primary immunodeficiencies and CLL, we conclude that the presence of pan-humoral immunodeficiency could itself be a risk factor for developing a CD8+ lymphogranulomatous reaction. This report and associated discussion evince that CD8+ predominant granulomatous reactions, distinct from sarcoidosis, may represent a previously unappreciated segment of the paraneoplastic granulomas observed in hematologic malignancies.


Sujet(s)
Lymphocytes T CD8+/anatomopathologie , Dermatite/immunologie , Granulome/immunologie , Sujet immunodéprimé , Leucémie chronique lymphocytaire à cellules B/complications , Syndromes paranéoplasiques/immunologie , Sujet âgé , Dermatite/anatomopathologie , Granulome/anatomopathologie , Humains , Mâle , Syndromes paranéoplasiques/anatomopathologie
14.
PLoS Pathog ; 17(10): e1009693, 2021 10.
Article de Anglais | MEDLINE | ID: mdl-34699567

RÉSUMÉ

Innate lymphoid cells (ILCs) comprise a heterogeneous population of immune cells that maintain barrier function and can initiate a protective or pathological immune response upon infection. Here we show the involvement of IL-17A-producing ILCs in microbiota-driven immunopathology in cutaneous leishmaniasis. IL-17A-producing ILCs were RORγt+ and were enriched in Leishmania major infected skin, and topical colonization with Staphylococcus epidermidis before L. major infection exacerbated the skin inflammatory responses and IL-17A-producing RORγt+ ILC accumulation without impacting type 1 immune responses. IL-17A responses in ILCs were directed by Batf3 dependent CD103+ dendritic cells and IL-23. Moreover, experiments using Rag1-/- mice established that IL-17A+ ILCs were sufficient in driving the inflammatory responses as depletion of ILCs or neutralization of IL-17A diminished the microbiota mediated immunopathology. Taken together, this study indicates that the skin microbiota promotes RORγt+ IL-17A-producing ILCs, which augment the skin inflammation in cutaneous leishmaniasis.


Sujet(s)
Cellules dendritiques/immunologie , Interleukine-17/immunologie , Leishmaniose cutanée/immunologie , Lymphocytes/immunologie , Peau/microbiologie , Animaux , Dermatite/immunologie , Dermatite/microbiologie , Immunité innée/immunologie , Leishmaniose cutanée/microbiologie , Souris
15.
EMBO J ; 40(22): e108234, 2021 11 15.
Article de Anglais | MEDLINE | ID: mdl-34586646

RÉSUMÉ

DNA methylation is a fundamental epigenetic modification, important across biological processes. The maintenance methyltransferase DNMT1 is essential for lineage differentiation during development, but its functions in tissue homeostasis are incompletely understood. We show that epidermis-specific DNMT1 deletion severely disrupts epidermal structure and homeostasis, initiating a massive innate immune response and infiltration of immune cells. Mechanistically, DNA hypomethylation in keratinocytes triggered transposon derepression, mitotic defects, and formation of micronuclei. DNA release into the cytosol of DNMT1-deficient keratinocytes activated signaling through cGAS and STING, thus triggering inflammation. Our findings show that disruption of a key epigenetic mark directly impacts immune and tissue homeostasis, and potentially impacts our understanding of autoinflammatory diseases and cancer immunotherapy.


Sujet(s)
Méthylation de l'ADN , Dermatite/génétique , Épiderme/physiopathologie , Nucleotidyltransferases/métabolisme , Protéines adaptatrices de la transduction du signal/métabolisme , Animaux , Aberrations des chromosomes , Cytosol/physiologie , DNA (Cytosine-5-)-methyltransferase 1/génétique , Dermatite/immunologie , Dermatite/anatomopathologie , Humains , Immunité innée/génétique , Hélicase IFIH1 inductrice de l'interféron/métabolisme , Kératinocytes/immunologie , Kératinocytes/métabolisme , Kératinocytes/anatomopathologie , Protéines membranaires/génétique , Protéines membranaires/métabolisme , Souris transgéniques , Nucleotidyltransferases/génétique
16.
J Am Acad Dermatol ; 85(5): 1274-1284, 2021 Nov.
Article de Anglais | MEDLINE | ID: mdl-34363909

RÉSUMÉ

Dermatologists diagnose and treat many immune-mediated inflammatory diseases (IMID). Understanding the inherent immune dysregulation of these diseases as well as the additional disruption that comes as a result of IMID treatments has been important during the COVID-19 pandemic. With vaccines becoming widely available, dermatologists need to be familiar with the risks and benefits of vaccination in these patients, particularly those taking biologics, in order to have informed discussions with their patients. In this review, we present the current evidence related to COVID-19 vaccine safety and efficacy in patients with IMID and review existing recommendations for vaccination against SARS-CoV-2. Given the current evidence, there is minimal concern that these patients are at any greater risk of harm from COVID-19 vaccination compared to healthy controls. For most, the benefit of avoiding severe COVID-19 through vaccination will outweigh the theoretical risk of these vaccines. A question that is still outstanding is whether patients on biologics will generate a sufficient immune response to the vaccine, which may be dependent on the specific biologic therapy and indication being treated. This underscores the importance of following patients with IMID after vaccination to determine the safety, efficacy, and duration of the vaccine in this population.


Sujet(s)
Vaccins contre la COVID-19/administration et posologie , Vaccins contre la COVID-19/effets indésirables , COVID-19/prévention et contrôle , Dermatite/immunologie , Sujet immunodéprimé , Produits biologiques/usage thérapeutique , Contre-indications aux médicaments , Dermatite/traitement médicamenteux , Humains , Facteurs immunologiques/usage thérapeutique , SARS-CoV-2
17.
Int J Mol Sci ; 22(14)2021 Jul 12.
Article de Anglais | MEDLINE | ID: mdl-34299063

RÉSUMÉ

Regulatory T cells (Tregs) suppress immune responses and maintain immunological self-tolerance and homeostasis. We currently investigated relationships between skin barrier condition and Treg behavior using skin barrier-disrupted mice. Skin barrier disruption was induced by repeated topical application of 4% sodium dodecyl sulfate (SDS) on mice. The number of CD4+ forkhead box protein P3 (Foxp3)+ Tregs was higher in 4% SDS-treated skins than in controls. This increasing was correlated with the degree of acanthosis. The numbers of interleukin (IL)-10+ and transforming growth factor (TGF)-ß+ Tregs also increased in 4% SDS-treated skins. Localization of IL-33 in keratinocytes shifted from nucleus to cytoplasm after skin barrier disruption. Notably, IL-33 promoted the migration of Tregs in chemotaxis assay. The skin infiltration of Tregs was cancelled in IL-33 neutralizing antibody-treated mice and IL-33 knockout mice. Thus, keratinocyte-derived IL-33 may induce Treg migration into barrier-disrupted skin to control the phase transition between healthy and inflammatory conditions.


Sujet(s)
Mouvement cellulaire , Chimiotaxie , Dermatite/anatomopathologie , Interleukine-33/physiologie , Peau/anatomopathologie , Lymphocytes T régulateurs/immunologie , Animaux , Dermatite/immunologie , Dermatite/métabolisme , Facteurs de transcription Forkhead/métabolisme , Mâle , Souris , Souris de lignée C57BL , Souris knockout , Peau/métabolisme
18.
Mediators Inflamm ; 2021: 6627087, 2021.
Article de Anglais | MEDLINE | ID: mdl-34257517

RÉSUMÉ

BACKGROUND: Immunoglobulin E (IgE) is the most important promoter of allergic inflammation. However, there are few systematic studies on IgE in age range, genders, disease spectrum, and time regularity. AIM: To screen the common allergens, allergen spectrum, and IgE difference between type 2 inflammatory allergic diseases and other allergic diseases in Weifang, China. METHODS: A retrospective study was performed by estimating patients' clinical data suffering from allergic diseases (urticaria, pollinosis, allergic rhinitis, atopic dermatitis, and bronchial asthma) between May 2019 and April 2020 using an allergen detection kit of Macro-Union Pharmaceutical. RESULTS: 732 of the 1367 patients showed different antigen positive, and the positive rate was 53.5%. The most common allergens were dust mites, mixed fungi, Artemisia pollen, cat/dog dander, and cockroaches. There were 27.0% (369/1367) of the patients with single positive allergen-specific IgE (sIgE), 26.5% (363/1367) with multiple-positive IgE. The total immunoglobulin E (tIgE) levels varied with gender, age, and type of disease. There was a difference in the distribution of allergens between children and adults. A positive correlation between the serum-specific IgE and the corresponding local inhaled allergen density was observed. CONCLUSIONS: In this study, we found that type 2 inflammatory allergic diseases have higher serum IgE and a higher probability of inhaled sIgE positive. According to age, gender, and condition, serological IgE detection of allergens provides new insight into the early diagnosis and prevention of allergic diseases.


Sujet(s)
Asthme/sang , Dermatite/sang , Hypersensibilité/sang , Immunoglobuline E/sang , Rhinite/sang , Adolescent , Adulte , Sujet âgé , Allergènes/sang , Asthme/immunologie , Enfant , Enfant d'âge préscolaire , Chine/épidémiologie , Dermatite/immunologie , Femelle , Humains , Hypersensibilité/immunologie , Nourrisson , Nouveau-né , Inflammation , Mâle , Adulte d'âge moyen , Reproductibilité des résultats , Études rétrospectives , Rhinite/immunologie , Jeune adulte
19.
Sci Rep ; 11(1): 9971, 2021 05 11.
Article de Anglais | MEDLINE | ID: mdl-33976265

RÉSUMÉ

Previously, we had cross-sectionally explored the characteristics of T cell receptor (TCR) repertoires from occupational medicamentosa-like dermatitis due to trichloroethylene (OMDT) patients, now we further analyzed the dynamic features of OMDT TCR repertoires. Peripheral blood TCR ß-chain complementarity-determining region 3 (CDR3) genes were detected with the high throughput sequencing in 24 OMDT cases in their acute, chronic and recovery stages, respectively, and in 24 trichloroethylene-exposed healthy controls. The TCR repertoire diversity, TRBV/TRBD/TRBJ gene usage and combination, frequencies of CDR3 nucleotide (nt) and amino acid (aa) sequences in the cases in different stages and in the controls were analyzed. TRBV6-4 and TRBV7-9 frequencies significantly differed between the cases and controls (both P < 6.1 × 10-4). TRBV6-4 combination with TRBJ2-1, TRBJ2-2, TRBJ2-3, and TRBJ2-6, and TRBV7-9 combination with TRBJ2-1 were associated with the stage by OMDT severity (all P < 0.001). Ten CDR3-nt and 7 CDR3-aa sequences in TRBV7-9-TRBJ2-1 combination and 1 CDR3-nt and 1 CDR3-aa sequences in TRBV6-4-TRBJ2-1 combination were identified as associated with the severity of OMDT (all P < 0.001). We revealed further how TCR repertoires vary with the severity in the development of OMDT, and severity-related TCRs may provide important therapeutic targets for OMDT in clinical practice.


Sujet(s)
Régions déterminant la complémentarité/métabolisme , Dermatite/immunologie , Maladies professionnelles/induit chimiquement , Récepteur lymphocytaire T antigène, alpha-bêta/métabolisme , Trichloroéthylène/toxicité , Adolescent , Adulte , Études cas-témoins , Femelle , Humains , Mâle , Maladies professionnelles/immunologie , Jeune adulte
20.
Int J Mol Sci ; 22(9)2021 Apr 27.
Article de Anglais | MEDLINE | ID: mdl-33925601

RÉSUMÉ

Mast cells (MCs) are best-known as key effector cells of immediate-type allergic reactions that may even culminate in life-threatening anaphylactic shock syndromes. However, strategically positioned at the host-environment interfaces and equipped with a plethora of receptors, MCs also play an important role in the first-line defense against pathogens. Their main characteristic, the huge amount of preformed proinflammatory mediators embedded in secretory granules, allows for a rapid response and initiation of further immune effector cell recruitment. The same mechanism, however, may account for detrimental overshooting responses. MCs are not only detrimental in MC-driven diseases but also responsible for disease exacerbation in other inflammatory disorders. Focusing on the skin as the largest immune organ, we herein review both beneficial and detrimental functions of skin MCs, from skin barrier integrity via host defense mechanisms to MC-driven inflammatory skin disorders. Moreover, we emphasize the importance of IgE-independent pathways of MC activation and their role in sustained chronic skin inflammation and disease exacerbation.


Sujet(s)
Mastocytes/immunologie , Mastocytes/métabolisme , Peau/immunologie , Anaphylaxie/immunologie , Animaux , Dermatite/immunologie , Humains , Inflammation/immunologie , Vésicules de sécrétion/immunologie , Vésicules de sécrétion/métabolisme , Peau/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE