Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 10 de 10
Filtrer
Plus de filtres











Base de données
Gamme d'année
1.
Immunity ; 56(6): 1239-1254.e7, 2023 06 13.
Article de Anglais | MEDLINE | ID: mdl-37028427

RÉSUMÉ

Early-life establishment of tolerance to commensal bacteria at barrier surfaces carries enduring implications for immune health but remains poorly understood. Here, we showed that tolerance in skin was controlled by microbial interaction with a specialized subset of antigen-presenting cells. More particularly, CD301b+ type 2 conventional dendritic cells (DCs) in neonatal skin were specifically capable of uptake and presentation of commensal antigens for the generation of regulatory T (Treg) cells. CD301b+ DC2 were enriched for phagocytosis and maturation programs, while also expressing tolerogenic markers. In both human and murine skin, these signatures were reinforced by microbial uptake. In contrast to their adult counterparts or other early-life DC subsets, neonatal CD301b+ DC2 highly expressed the retinoic-acid-producing enzyme, RALDH2, the deletion of which limited commensal-specific Treg cell generation. Thus, synergistic interactions between bacteria and a specialized DC subset critically support early-life tolerance at the cutaneous interface.


Sujet(s)
Cellules dendritiques , Peau , Animaux , Souris , Humains , Lymphocytes T régulateurs , Tolérance immunitaire , Aldehyde oxidoreductases/métabolisme
2.
J Invest Dermatol ; 143(5): 790-800.e12, 2023 05.
Article de Anglais | MEDLINE | ID: mdl-36496196

RÉSUMÉ

FLG variants underlie ichthyosis vulgaris and increased risk of atopic dermatitis, conditions typified by disruption of the skin microbiome and cutaneous immune response. Yet, it remains unclear whether neonatal skin barrier compromise because of FLG deficiency alters the quality of commensal-specific T cells and the functional impact of such responses. To address these questions, we profiled changes in the skin barrier and early cutaneous immune response of neonatal C57BL/6 Flg‒/‒ and wild-type mice using single-cell RNA sequencing, flow cytometry, and other modalities. Flg‒/‒ neonates showed little alteration in transepidermal water loss or lipid- or corneocyte-related gene expression. However, they showed increases in barrier disruption genes, epidermal dye penetration, and numbers of skin CD4+ T cells. Using an engineered strain of Staphylococcus epidermidis (S. epidermidis 2W) to study the response to neonatal skin colonization, we found that commensal-specific CD4+ T cells were skewed in Flg‒/‒ pups toward effector rather than regulatory T cells. This altered response persisted into adulthood, where it was typified by T helper 17 (Th17) cells and associated with increased susceptibility to imiquimod-induced skin inflammation. Thus, subtle but impactful differences in neonatal barrier function in Flg‒/‒ mice are accompanied by a skewed commensal-specific CD4+ response, with enduring consequences for skin immune homeostasis.


Sujet(s)
Eczéma atopique , Protéines de filaments intermédiaires , Animaux , Souris , Bactéries , Lymphocytes T CD4+ , Eczéma atopique/génétique , Protéines de filaments intermédiaires/génétique , Souris de lignée C57BL , Peau
3.
Cell Rep ; 39(9): 110891, 2022 05 31.
Article de Anglais | MEDLINE | ID: mdl-35649365

RÉSUMÉ

Resident microbes in skin and gut predominantly impact local immune cell function during homeostasis. However, colitis-associated neutrophilic skin disorders suggest possible breakdown of this compartmentalization with disease. Using a model wherein neonatal skin colonization by Staphylococcus epidermidis facilitates generation of commensal-specific tolerance and CD4+ regulatory T cells (Tregs), we ask whether this response is perturbed by gut inflammation. Chemically induced colitis is accompanied by intestinal expansion of S. epidermidis and reduces gut-draining lymph node (dLN) commensal-specific Tregs. It also results in reduced commensal-specific Tregs in skin and skin-dLNs and increased skin neutrophils. Increased CD4+ circulation between gut and skin dLN suggests that the altered cutaneous response is initiated in the colon, and resistance to colitis-induced effects in Cd4creIl1r1fl/fl mice implicate interleukin (IL)-1 in mediating the altered commensal-specific response. These findings provide mechanistic insight into observed connections between inflammatory skin and intestinal diseases.


Sujet(s)
Colite , Immunité , Animaux , Colite/induit chimiquement , Inflammation , Souris , Peau , Staphylococcus epidermidis , Lymphocytes T régulateurs
4.
Cell Host Microbe ; 29(5): 742-744, 2021 05 12.
Article de Anglais | MEDLINE | ID: mdl-33984276

RÉSUMÉ

Microbes can boost cutaneous immune defense and skin reparative capacity. However, mechanistic understanding, especially of the latter, remains sparse. In this issue of Cell Host & Microbe, Wang et al. (2021) shed light on this, demonstrating that bacteria contribute to hair follicle neogenesis after skin wounding via keratinocyte-intrinsic IL-1R1 signaling.


Sujet(s)
Follicule pileux , Peau , Animaux , Bactéries , Souris , Régénération , Transduction du signal
5.
Clin Exp Allergy ; 50(8): 904-914, 2020 08.
Article de Anglais | MEDLINE | ID: mdl-32542913

RÉSUMÉ

BACKGROUND: Sialic acid-binding immunoglobulin-like lectin (Siglec)-8 is expressed on mast cells and eosinophils, but information about Siglec-8 expression and function in the lung is limited. A humanized antibody, AK002, targeting Siglec-8 is undergoing development for treatment of diseases associated with mast cell and eosinophil-driven inflammation. OBJECTIVE: To characterize Siglec-8 expression in the airway in asthma and determine whether antibodies that target Siglec-8 (S8mAbs) can decrease airway eosinophils in asthma or inhibit lung mast cell activation. METHODS: Gene expression profiling and flow cytometry were used to characterize Siglec-8 expression in sputum cells from stable asthma. An antibody-dependent cellular cytotoxicity (ADCC) assay was used to determine whether an S8mAb can decrease eosinophils in sputum from asthma patients ex vivo. A mast cell activation assay was used to determine whether an S8mAb can inhibit mast cell activation in human lung tissue ex vivo. RESULTS: Gene expression for Siglec-8 is increased in sputum cells in asthma and correlates with gene expression for eosinophils and mast cells. Gene expression for Siglec-8 is inversely and significantly correlated with measures of airflow obstruction in asthma patients. Siglec-8 is prominently expressed on the surface of eosinophils and mast cells in sputum. S8mAbs decrease eosinophils in sputum from patients with asthma and inhibit FcεR1-activated mast cells in lung tissues. CONCLUSIONS AND CLINICAL RELEVANCE: Siglec-8 is highly expressed on eosinophils and mast cells in asthmatic sputum and targeting Siglec-8 with an antibody is a plausible strategy to decrease sputum eosinophils and inhibit lung mast cells in asthma.


Sujet(s)
Antiasthmatiques/pharmacologie , Anticorps monoclonaux humanisés/pharmacologie , Asthme/traitement médicamenteux , Granulocytes éosinophiles/effets des médicaments et des substances chimiques , Lectines/antagonistes et inhibiteurs , Poumon/effets des médicaments et des substances chimiques , Mastocytes/effets des médicaments et des substances chimiques , Adulte , Cytotoxicité à médiation cellulaire dépendante des anticorps/effets des médicaments et des substances chimiques , Antigènes CD/génétique , Antigènes CD/immunologie , Antigènes CD/métabolisme , Antigènes de différenciation des lymphocytes B/génétique , Antigènes de différenciation des lymphocytes B/immunologie , Antigènes de différenciation des lymphocytes B/métabolisme , Apoptose/effets des médicaments et des substances chimiques , Asthme/immunologie , Asthme/métabolisme , Études cas-témoins , Cellules cultivées , Granulocytes éosinophiles/immunologie , Granulocytes éosinophiles/métabolisme , Femelle , Humains , Lectines/génétique , Lectines/immunologie , Lectines/métabolisme , Poumon/immunologie , Poumon/métabolisme , Mâle , Mastocytes/immunologie , Mastocytes/métabolisme , Adulte d'âge moyen , Récepteurs aux IgE/génétique , Récepteurs aux IgE/métabolisme , Expectoration/cytologie , Jeune adulte
7.
Cell Host Microbe ; 26(6): 795-809.e5, 2019 12 11.
Article de Anglais | MEDLINE | ID: mdl-31784259

RÉSUMÉ

The host must develop tolerance to commensal microbes and protective responses to infectious pathogens, yet the mechanisms enabling a privileged relationship with commensals remain largely unknown. Skin colonization by commensal Staphylococcus epidermidis facilitates immune tolerance preferentially in neonates via induction of antigen-specific regulatory T cells (Tregs). Here, we demonstrate that this tolerance is not indiscriminately extended to all bacteria encountered in this early window. Rather, neonatal colonization by Staphylococcus aureus minimally enriches for antigen-specific Tregs and does not prevent skin inflammation upon later-life exposure. S. aureus α-toxin contributes to this response by stimulating myeloid cell production of IL-1ß, which limits S. aureus-specific Tregs. Loss of α-toxin or the IL-1 receptor increases Treg enrichment, whereas topical application of IL-1ß or α-toxin diminishes tolerogenic responses to S. epidermidis. Thus, the preferential activation of a key alarmin pathway facilitates early discrimination of microbial "foe" from "friend," thereby preventing tolerance to a common skin pathogen.


Sujet(s)
Toxines bactériennes/immunologie , Récepteurs à l'interleukine-1/métabolisme , Peau/microbiologie , Infections à staphylocoques/immunologie , Lymphocytes T régulateurs/immunologie , Animaux , Animaux nouveau-nés , Toxines bactériennes/métabolisme , Interactions hôte-microbes/immunologie , Tolérance immunitaire , Souris , Récepteurs à l'interleukine-1/immunologie , Transduction du signal/immunologie , Staphylococcus aureus/immunologie , Staphylococcus epidermidis/immunologie , Symbiose/immunologie , Virulence/immunologie
8.
Proc Natl Acad Sci U S A ; 113(31): 8765-70, 2016 08 02.
Article de Anglais | MEDLINE | ID: mdl-27432971

RÉSUMÉ

Type 2 inflammation occurs in a large subgroup of asthmatics, and novel cytokine-directed therapies are being developed to treat this population. In mouse models, interleukin-33 (IL-33) activates lung resident innate lymphoid type 2 cells (ILC2s) to initiate airway type 2 inflammation. In human asthma, which is chronic and difficult to model, the role of IL-33 and the target cells responsible for persistent type 2 inflammation remain undefined. Full-length IL-33 is a nuclear protein and may function as an "alarmin" during cell death, a process that is uncommon in chronic stable asthma. We demonstrate a previously unidentified mechanism of IL-33 activity that involves alternative transcript splicing, which may operate in stable asthma. In human airway epithelial cells, alternative splicing of the IL-33 transcript is consistently present, and the deletion of exons 3 and 4 (Δ exon 3,4) confers cytoplasmic localization and facilitates extracellular secretion, while retaining signaling capacity. In nonexacerbating asthmatics, the expression of Δ exon 3,4 is strongly associated with airway type 2 inflammation, whereas full-length IL-33 is not. To further define the extracellular role of IL-33 in stable asthma, we sought to determine the cellular targets of its activity. Comprehensive flow cytometry and RNA sequencing of sputum cells suggest basophils and mast cells, not ILC2s, are the cellular sources of type 2 cytokines in chronic asthma. We conclude that IL-33 isoforms activate basophils and mast cells to drive type 2 inflammation in chronic stable asthma, and novel IL-33 inhibitors will need to block all biologically active isoforms.


Sujet(s)
Épissage alternatif , Asthme/génétique , Inflammation/génétique , Interleukine-33/génétique , Adulte , Asthme/métabolisme , Granulocytes basophiles/métabolisme , Lignée cellulaire , Cellules épithéliales/métabolisme , Femelle , Analyse de profil d'expression de gènes/méthodes , Humains , Inflammation/métabolisme , Protéine-1 analogue au récepteur de l'interleukin-1/génétique , Protéine-1 analogue au récepteur de l'interleukin-1/métabolisme , Interleukine-33/métabolisme , Mâle , Mastocytes/métabolisme , Adulte d'âge moyen , Muqueuse respiratoire/métabolisme , Muqueuse respiratoire/anatomopathologie , Expectoration/cytologie , Expectoration/métabolisme , Jeune adulte
9.
Immunity ; 44(4): 821-32, 2016 Apr 19.
Article de Anglais | MEDLINE | ID: mdl-26850657

RÉSUMÉ

MicroRNAs (miRNAs) are important regulators of cell fate decisions in immune responses. They act by coordinate repression of multiple target genes, a property that we exploited to uncover regulatory networks that govern T helper-2 (Th2) cells. A functional screen of individual miRNAs in primary T cells uncovered multiple miRNAs that inhibited Th2 cell differentiation. Among these were miR-24 and miR-27, miRNAs coexpressed from two genomic clusters, which each functioned independently to limit interleukin-4 (IL-4) production. Mice lacking both clusters in T cells displayed increased Th2 cell responses and tissue pathology in a mouse model of asthma. Gene expression and pathway analyses placed miR-27 upstream of genes known to regulate Th2 cells. They also identified targets not previously associated with Th2 cell biology which regulated IL-4 production in unbiased functional testing. Thus, elucidating the biological function and target repertoire of miR-24 and miR-27 reveals regulators of Th2 cell biology.


Sujet(s)
Asthme/immunologie , Interleukine-4/biosynthèse , microARN/génétique , Lymphocytes auxiliaires Th2/immunologie , Animaux , Séquence nucléotidique , Différenciation cellulaire/génétique , Différenciation cellulaire/immunologie , Cellules cultivées , Modèles animaux de maladie humaine , Femelle , Inflammation/immunologie , Interleukine-4/immunologie , Activation des lymphocytes/immunologie , Mâle , Souris , Souris knockout , Famille multigénique/génétique , Analyse de séquence d'ARN , Lymphocytes auxiliaires Th2/cytologie
10.
Elife ; 42015 Nov 13.
Article de Anglais | MEDLINE | ID: mdl-26565589

RÉSUMÉ

Recognition and elimination of tumor cells by the immune system is crucial for limiting tumor growth. Natural killer (NK) cells become activated when the receptor NKG2D is engaged by ligands that are frequently upregulated in primary tumors and on cancer cell lines. However, the molecular mechanisms driving NKG2D ligand expression on tumor cells are not well defined. Using a forward genetic screen in a tumor-derived human cell line, we identified several novel factors supporting expression of the NKG2D ligand ULBP1. Our results show stepwise contributions of independent pathways working at multiple stages of ULBP1 biogenesis. Deeper investigation of selected hits from the screen showed that the transcription factor ATF4 drives ULBP1 gene expression in cancer cell lines, while the RNA-binding protein RBM4 supports ULBP1 expression by suppressing a novel alternatively spliced isoform of ULBP1 mRNA. These findings offer insight into the stress pathways that alert the immune system to danger.


Sujet(s)
Protéines et peptides de signalisation intracellulaire/métabolisme , Cellules tueuses naturelles/immunologie , Activation des lymphocytes , Facteur de transcription ATF-4/métabolisme , Lignée cellulaire tumorale , Protéines liées au GPI/génétique , Protéines liées au GPI/métabolisme , Dépistage génétique , Humains , Protéines et peptides de signalisation intracellulaire/génétique , Sous-famille K des récepteurs de cellules NK de type lectine/métabolisme , Protéines de liaison à l'ARN/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE