Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 3 de 3
Filtrer
Plus de filtres










Base de données
Gamme d'année
1.
Cell Rep Med ; 1(8): 100132, 2020 11 17.
Article de Anglais | MEDLINE | ID: mdl-33294857

RÉSUMÉ

Lymphocytes in barrier tissues play critical roles in host defense and homeostasis. These cells take up residence in tissues during defined developmental windows, when they may demonstrate distinct phenotypes and functions. Here, we utilized mass and flow cytometry to elucidate early features of human skin immunity. Although most conventional αß T (Tconv) cells in fetal skin have a naive, proliferative phenotype, a subset of CD4+ Tconv and CD8+ cells demonstrate memory-like features and a propensity for interferon (IFN)γ production. Skin regulatory T cells dynamically accumulate over the second trimester in temporal and regional association with hair follicle development. These fetal skin regulatory T cells (Tregs) demonstrate an effector memory phenotype while differing from their adult counterparts in expression of key effector molecules. Thus, we identify features of prenatal skin lymphocytes that may have key implications for understanding antigen and allergen encounters in utero and in infancy.


Sujet(s)
Lymphocytes T CD4+/immunologie , Lymphocytes T CD8+/immunologie , Mémoire immunologique/immunologie , Interféron gamma/immunologie , Peau/immunologie , Cytométrie en flux/méthodes , Humains , Activation des lymphocytes/immunologie , Lymphocytes T régulateurs/immunologie
2.
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
3.
Cell Host Microbe ; 21(4): 467-477.e5, 2017 Apr 12.
Article de Anglais | MEDLINE | ID: mdl-28343820

RÉSUMÉ

Regulatory T cells (Tregs) are required to establish immune tolerance to commensal microbes. Tregs accumulate abruptly in the skin during a defined window of postnatal tissue development. However, the mechanisms mediating Treg migration to neonatal skin are unknown. Here we show that hair follicle (HF) development facilitates the accumulation of Tregs in neonatal skin and that upon skin entry these cells localize to HFs, a primary reservoir for skin commensals. Further, germ-free neonates had reduced skin Tregs indicating that commensal microbes augment Treg accumulation. We identified Ccl20 as a HF-derived, microbiota-dependent chemokine and found its receptor, Ccr6, to be preferentially expressed by Tregs in neonatal skin. The Ccl20-Ccr6 pathway mediated Treg migration in vitro and in vivo. Thus, HF morphogenesis, commensal microbe colonization, and local chemokine production work in concert to recruit Tregs into neonatal skin, thereby establishing this tissue Treg niche early in life.


Sujet(s)
Follicule pileux/croissance et développement , Microbiote/immunologie , Morphogenèse , Peau/immunologie , Peau/microbiologie , Symbiose , Lymphocytes T régulateurs/immunologie , Animaux , Chimiokine CCL20/métabolisme , Follicule pileux/immunologie , Follicule pileux/microbiologie , Tolérance immunitaire , Souris , Récepteurs CCR6/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...