Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Immunity ; 56(6): 1239-1254.e7, 2023 06 13.
Artículo en Inglés | MEDLINE | ID: mdl-37028427

RESUMEN

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.


Asunto(s)
Células Dendríticas , Piel , Animales , Ratones , Humanos , Linfocitos T Reguladores , Tolerancia Inmunológica , Aldehído Oxidorreductasas/metabolismo
2.
Cell ; 169(6): 1119-1129.e11, 2017 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-28552347

RESUMEN

The maintenance of tissue homeostasis is critically dependent on the function of tissue-resident immune cells and the differentiation capacity of tissue-resident stem cells (SCs). How immune cells influence the function of SCs is largely unknown. Regulatory T cells (Tregs) in skin preferentially localize to hair follicles (HFs), which house a major subset of skin SCs (HFSCs). Here, we mechanistically dissect the role of Tregs in HF and HFSC biology. Lineage-specific cell depletion revealed that Tregs promote HF regeneration by augmenting HFSC proliferation and differentiation. Transcriptional and phenotypic profiling of Tregs and HFSCs revealed that skin-resident Tregs preferentially express high levels of the Notch ligand family member, Jagged 1 (Jag1). Expression of Jag1 on Tregs facilitated HFSC function and efficient HF regeneration. Taken together, our work demonstrates that Tregs in skin play a major role in HF biology by promoting the function of HFSCs.


Asunto(s)
Folículo Piloso/citología , Células Madre/metabolismo , Linfocitos T Reguladores/metabolismo , Animales , Células Epiteliales/metabolismo , Folículo Piloso/metabolismo , Humanos , Inflamación/metabolismo , Proteína Jagged-1/metabolismo , Ratones
3.
Immunity ; 55(9): 1586-1588, 2022 09 13.
Artículo en Inglés | MEDLINE | ID: mdl-36103856

RESUMEN

The full range of receptors through which antimicrobial peptides exert their immunologic effects remains incompletely explored. Dong and colleagues identify Mgrpra2 as a G-coupled protein receptor on neutrophils, for which keratinocyte-derived Beta-defensins serve as key ligands. Binding of Mgrpra2 leads to release of neutrophil granules and Il-1ß, which helps shape skin microbiome composition and augments cutaneous defense against bacterial infection.


Asunto(s)
beta-Defensinas , Proteínas Portadoras , Queratinocitos/metabolismo , Neutrófilos/metabolismo , Piel/metabolismo , beta-Defensinas/química , beta-Defensinas/metabolismo
4.
Artículo en Inglés | MEDLINE | ID: mdl-38734386

RESUMEN

BACKGROUND: The contribution of Staphylococcus aureus to the exacerbation of atopic dermatitis (AD) is widely documented, but its role as a primary trigger of AD skin symptoms remains poorly explored. OBJECTIVES: This study sought to reappraise the main bacterial factors and underlying immune mechanisms by which S aureus triggers AD-like inflammation. METHODS: This study capitalized on a preclinical model, in which different clinical isolates were applied in the absence of any prior experimental skin injury. RESULTS: The development of S aureus-induced dermatitis depended on the nature of the S aureus strain, its viability, the concentration of the applied bacterial suspension, the production of secreted and nonsecreted factors, as well as the activation of accessory gene regulatory quorum sensing system. In addition, the rising dermatitis, which exhibited the well-documented AD cytokine signature, was significantly inhibited in inflammasome adaptor apoptosis-associated speck-like protein containing a CARD domain- and monocyte/macrophage-deficient animals, but not in T- and B-cell-deficient mice, suggesting a major role for the innate response in the induction of skin inflammation. However, bacterial exposure generated a robust adaptive immune response against S aureus, and an accumulation of S aureus-specific γδ and CD4+ tissue resident memory T cells at the site of previous dermatitis. The latter both contributed to worsen the flares of AD-like dermatitis on new bacteria exposures, but also, protected the mice from persistent bacterial colonization. CONCLUSIONS: These data highlight the induction of unique AD-like inflammation, with the generation of proinflammatory but protective tissue resident memory T cells in a context of natural exposure to pathogenic S aureus strains.

5.
Trends Immunol ; 42(12): 1088-1099, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34743922

RESUMEN

Early life is a dynamic period for skin microbial colonization and immune development. We postulate that microbial exposures in this period durably alter the skin immune trajectory and later disease susceptibility. Bacteria contribute to infant skin immune imprinting via interactions with microbes as well as with cutaneous epithelial and immune cells. Excellent research is underway at the skin microbiome-immune interface, both in deciphering basic mechanisms and implementing their therapeutic applications. As emphasized herein, focusing on the unique opportunities and challenges presented by microbial immune modulation in early life will be important. In our view, only through dedicated study of skin-microbe crosstalk in this developmental window can we elucidate the molecular underpinnings of pivotal events that contribute to sustained host-microbe symbiosis.


Asunto(s)
Microbiota , Bacterias , Humanos , Lactante , Piel/microbiología , Simbiosis
6.
Immunity ; 43(5): 1011-21, 2015 Nov 17.
Artículo en Inglés | MEDLINE | ID: mdl-26588783

RESUMEN

The skin is a site of constant dialog between the immune system and commensal bacteria. However, the molecular mechanisms that allow us to tolerate the presence of skin commensals without eliciting destructive inflammation are unknown. Using a model system to study the antigen-specific response to S. epidermidis, we demonstrated that skin colonization during a defined period of neonatal life was required for establishing immune tolerance to commensal microbes. This crucial window was characterized by an abrupt influx of highly activated regulatory T (Treg) cells into neonatal skin. Selective inhibition of this Treg cell wave completely abrogated tolerance. Thus, the host-commensal relationship in the skin relied on a unique Treg cell population that mediated tolerance to bacterial antigens during a defined developmental window. This suggests that the cutaneous microbiome composition in neonatal life is crucial in shaping adaptive immune responses to commensals, and disrupting these interactions might have enduring health implications.


Asunto(s)
Animales Recién Nacidos/inmunología , Piel/inmunología , Piel/microbiología , Infecciones Estafilocócicas/inmunología , Staphylococcus epidermidis/inmunología , Linfocitos T Reguladores/inmunología , Animales , Animales Recién Nacidos/microbiología , Antígenos Bacterianos/inmunología , Interacciones Huésped-Patógeno/inmunología , Tolerancia Inmunológica/inmunología , Inflamación/inmunología , Inflamación/microbiología , Ratones , Ratones Endogámicos C57BL , Microbiota/inmunología , Datos de Secuencia Molecular , Infecciones Estafilocócicas/microbiología , Linfocitos T Reguladores/microbiología
7.
J Immunol ; 207(7): 1763-1775, 2021 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-34470859

RESUMEN

Regulatory T cells (Tregs) reside in nonlymphoid tissues where they carry out unique functions. The molecular mechanisms responsible for Treg accumulation and maintenance in these tissues are relatively unknown. Using an unbiased discovery approach, we identified LAYN (layilin), a C-type lectin-like receptor, to be preferentially and highly expressed on a subset of activated Tregs in healthy and diseased human skin. Expression of layilin on Tregs was induced by TCR-mediated activation in the presence of IL-2 or TGF-ß. Mice with a conditional deletion of layilin in Tregs had reduced accumulation of these cells in tumors. However, these animals somewhat paradoxically had enhanced immune regulation in the tumor microenvironment, resulting in increased tumor growth. Mechanistically, layilin expression on Tregs had a minimal effect on their activation and suppressive capacity in vitro. However, expression of this molecule resulted in a cumulative anchoring effect on Treg dynamic motility in vivo. Taken together, our results suggest a model whereby layilin facilitates Treg adhesion in skin and, in doing so, limits their suppressive capacity. These findings uncover a unique mechanism whereby reduced Treg motility acts to limit immune regulation in nonlymphoid organs and may help guide strategies to exploit this phenomenon for therapeutic benefit.


Asunto(s)
Proteínas Portadoras/metabolismo , Glicoproteínas de Membrana/metabolismo , Receptores Mensajeros de Linfocitos/metabolismo , Piel/inmunología , Linfocitos T Reguladores/inmunología , Animales , Proteínas Portadoras/genética , Movimiento Celular , Células Cultivadas , Humanos , Tolerancia Inmunológica , Activación de Linfocitos , Glicoproteínas de Membrana/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Modelos Inmunológicos , Especificidad de Órganos , Receptores Mensajeros de Linfocitos/genética , Factor de Crecimiento Transformador beta/metabolismo
8.
Trends Immunol ; 39(4): 259-261, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29478772

RESUMEN

Although our knowledge of host-commensal interactions has increased exponentially, the mechanisms linking a specific commensal, its detection by the immune system, and its impact on tissue function are still often poorly understood. In a recent study in Cell, Linehan et al. dissect one of these interactions in the context of the skin, and demonstrate that Staphylococcus epidermidis antigens, presented through a non-classical pathway, drive the accumulation of CD8+ T cells that promote wound healing.


Asunto(s)
Piel , Simbiosis , Linfocitos T CD8-positivos , Staphylococcus epidermidis
9.
J Allergy Clin Immunol ; 143(1): 26-35, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30476499

RESUMEN

As an interface with the environment, the skin is a complex ecosystem colonized by many microorganisms that coexist in an established balance. The cutaneous microbiome inhibits colonization with pathogens, such as Staphylococcus aureus, and is a crucial component for function of the epidermal barrier. Moreover, crosstalk between commensals and the immune system is now recognized because microorganisms can modulate both innate and adaptive immune responses. Host-commensal interactions also have an effect on the developing immune system in infants and, subsequently, the occurrence of diseases, such as asthma and atopic dermatitis (AD). Later in life, the cutaneous microbiome contributes to the development and course of skin disease. Accordingly, in patients with AD, a decrease in microbiome diversity correlates with disease severity and increased colonization with pathogenic bacteria, such as S aureus. Early clinical studies suggest that topical application of commensal organisms (eg, Staphylococcus hominis or Roseomonas mucosa) reduces AD severity, which supports an important role for commensals in decreasing S aureus colonization in patients with AD. Advancing knowledge of the cutaneous microbiome and its function in modulating the course of skin disorders, such as AD, might result in novel therapeutic strategies.


Asunto(s)
Inmunidad Adaptativa , Dermatitis Atópica , Inmunidad Innata , Microbiota/inmunología , Infecciones Cutáneas Estafilocócicas , Staphylococcus aureus/inmunología , Dermatitis Atópica/inmunología , Dermatitis Atópica/microbiología , Dermatitis Atópica/patología , Femenino , Humanos , Masculino , Piel/inmunología , Piel/microbiología , Piel/patología , Infecciones Cutáneas Estafilocócicas/inmunología , Infecciones Cutáneas Estafilocócicas/microbiología , Infecciones Cutáneas Estafilocócicas/patología
10.
J Immunol ; 196(5): 2010-4, 2016 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-26826250

RESUMEN

Foxp3-expressing regulatory T cells (Tregs) reside in tissues where they control inflammation and mediate tissue-specific functions. The skin of mice and humans contain a large number of Tregs; however, the mechanisms of how these cells function in skin remain largely unknown. In this article, we show that Tregs facilitate cutaneous wound healing. Highly activated Tregs accumulated in skin early after wounding, and specific ablation of these cells resulted in delayed wound re-epithelialization and kinetics of wound closure. Tregs in wounded skin attenuated IFN-γ production and proinflammatory macrophage accumulation. Upon wounding, Tregs induce expression of the epidermal growth factor receptor (EGFR). Lineage-specific deletion of EGFR in Tregs resulted in reduced Treg accumulation and activation in wounded skin, delayed wound closure, and increased proinflammatory macrophage accumulation. Taken together, our results reveal a novel role for Tregs in facilitating skin wound repair and suggest that they use the EGFR pathway to mediate these effects.


Asunto(s)
Receptores ErbB/inmunología , Linfocitos T Reguladores/inmunología , Cicatrización de Heridas/inmunología , Animales , Activación de Linfocitos/inmunología , Ratones , Ratones Endogámicos C57BL , Subgrupos de Linfocitos T/inmunología
12.
J Invest Dermatol ; 144(5): 1001-1009, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38573278

RESUMEN

Whereas clinically apparent atopic dermatitis (AD) can be confirmed by validated diagnostic criteria, the preclinical phenotype of infants who eventually develop AD is less well-characterized. Analogous to unaffected or nonlesional skin in established AD, clinically normal-appearing skin in infants who will develop clinical AD has distinct changes. Prospective studies have revealed insights into this preclinical AD phenotype. In this study, we review the structural, immunologic, and microbiome nature of the preclinical AD phenotype. Determination of markers that predict the development of AD will facilitate targeting of interventions to prevent the development or reduce the severity of AD in infants.


Asunto(s)
Dermatitis Atópica , Piel , Humanos , Lactante , Biomarcadores/análisis , Dermatitis Atópica/inmunología , Dermatitis Atópica/microbiología , Dermatitis Atópica/diagnóstico , Microbiota/inmunología , Fenotipo , Índice de Severidad de la Enfermedad , Piel/microbiología , Piel/inmunología , Piel/patología
13.
Drug Discov Today Dis Mech ; 10(3-4)2013 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-24273587

RESUMEN

Our skin is home to a rich community of microorganisms. Recent advances in sequencing technology have allowed more accurate enumeration of these human-associated microbiota and investigation of their genomic content. Staphylococcus, Corynebacterium and Propionibacterium represent the dominant bacterial genera on skin and illustrate how bacteria adapt to life in this harsh environment and also provide us with unique benefits. In healthy states, our skin peacefully co-exists with commensal bacteria while fending off potentially dangerous invaders. Disruption of this equilibrium, termed "dysbiosis", can result from changes in the composition of our skin bacteria, an altered immune response to them, or both and may be a driving factor in certain types of inflammatory skin disease. Engineering topical therapeutics to favourably influence the composition of our skin flora and optimize interactions with them represents a real therapeutic opportunity for the field of dermatology and warrants additional investigation into skin microbial ecology and disease mechanisms related to host-microbe dysbiosis.

14.
J Invest Dermatol ; 143(5): 790-800.e12, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-36496196

RESUMEN

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.


Asunto(s)
Dermatitis Atópica , Proteínas de Filamentos Intermediarios , Animales , Ratones , Bacterias , Linfocitos T CD4-Positivos , Dermatitis Atópica/genética , Proteínas de Filamentos Intermediarios/genética , Ratones Endogámicos C57BL , Piel
15.
bioRxiv ; 2023 Oct 02.
Artículo en Inglés | MEDLINE | ID: mdl-37873143

RESUMEN

Early life microbe-immune interactions at barrier surfaces have lasting impacts on the trajectory towards health versus disease. Monocytes, macrophages and dendritic cells are primary sentinels in barrier tissues, yet the salient contributions of commensal-myeloid crosstalk during tissue development remain poorly understood. Here, we identify that commensal microbes facilitate accumulation of a population of monocytes in neonatal skin. Transient postnatal depletion of these monocytes resulted in heightened IL-17A production by skin T cells, which was particularly sustained among CD4+ T cells into adulthood and sufficient to exacerbate inflammatory skin pathologies. Neonatal skin monocytes were enriched in expression of negative regulators of the IL-1 pathway. Functional in vivo experiments confirmed a key role for excessive IL-1R1 signaling in T cells as contributing to the dysregulated type 17 response in neonatal monocyte-depleted mice. Thus, a commensal-driven wave of monocytes into neonatal skin critically facilitates long-term immune homeostasis in this prominent barrier tissue.

16.
Cell Host Microbe ; 30(5): 684-695, 2022 05 11.
Artículo en Inglés | MEDLINE | ID: mdl-35550671

RESUMEN

Our skin is the interface through which we mediate lifelong interactions with our surrounding environment. Initial development of the skin's epidermis, adnexal structures, and barrier function is necessary for normal cutaneous microbial colonization, immune development, and prevention of disease. Early life microbial exposures can have unique and long-lasting impacts on skin health. The identity of neonatal skin microbes and the context in which they are first encountered, i.e., through a compromised skin barrier or in conjunction with cutaneous inflammation, can have additional short- and long-term health consequences. Here, we discuss key attributes of infant skin and endogenous and exogenous factors that shape its relationship to the early life cutaneous microbiome, with a focus on their clinical implications.


Asunto(s)
Dermatitis , Microbiota , Interacciones Microbiota-Huesped , Humanos , Lactante , Recién Nacido , Piel
17.
Mucosal Immunol ; 15(4): 551-561, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35361906

RESUMEN

Our skin contributes critically to health via its role as a barrier tissue, carefully regulating passage of key substrates while also providing defense against exogenous threats. Immunological processes are integral to almost every skin function and paramount to our ability to live symbiotically with skin commensal microbes and other environmental stimuli. While many parallels can be drawn to immunobiology at other mucosal sites, skin immunity demonstrates unique features that relate to its distinct topography, chemical composition and microbial ecology. Here we provide an overview of skin as an immune organ, with reference to the broader context of mucosal immunology. We review paradigms of innate as well as adaptive immune function and highlight how skin-specific structures such as hair follicles and sebaceous glands interact and contribute to these processes. Finally, we highlight for the mucosal immunology community a few emerging areas of interest for the skin immunity field moving forward.


Asunto(s)
Membrana Mucosa , Simbiosis , Biología , Inmunidad Innata
18.
Cell Rep ; 39(9): 110891, 2022 05 31.
Artículo en Inglés | MEDLINE | ID: mdl-35649365

RESUMEN

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.


Asunto(s)
Colitis , Inmunidad , Animales , Colitis/inducido químicamente , Inflamación , Ratones , Piel , Staphylococcus epidermidis , Linfocitos T Reguladores
19.
Cell Host Microbe ; 29(5): 742-744, 2021 05 12.
Artículo en Inglés | MEDLINE | ID: mdl-33984276

RESUMEN

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.


Asunto(s)
Folículo Piloso , Piel , Animales , Bacterias , Ratones , Regeneración , Transducción de Señal
20.
Sci Immunol ; 6(62)2021 08 27.
Artículo en Inglés | MEDLINE | ID: mdl-34452925

RESUMEN

Regulatory T cells (Tregs) use multiple mechanisms to attenuate inflammation and prevent autoimmunity. Tregs residing in peripheral (i.e., nonlymphoid) tissues have specialized functions; specifically, skin Tregs promote wound healing, suppress dermal fibrosis, facilitate epidermal regeneration, and augment hair follicle cycling. Here, we demonstrated that skin Tregs were transcriptionally attuned to interact with their tissue environment through increased expression of integrin and TGF-ß pathway genes that influence epithelial cell biology. We identified a molecular pathway where skin Tregs license keratinocytes to promote innate inflammation after skin barrier breach. Using a single-cell discovery approach, we identified preferential expression of the integrin αvß8 on skin Tregs Upon skin injury, Tregs used this integrin to activate latent TGF-ß, which acted directly on epithelial cells to promote CXCL5 production and neutrophil recruitment. Induction of this circuit delayed epidermal regeneration but provided protection from Staphylococcus aureus infection across a compromised barrier. Thus, αvß8-expressing Tregs in the skin, somewhat paradoxical to their canonical immunosuppressive functions, facilitated inflammation acutely after loss of barrier integrity to promote host defense against infection.


Asunto(s)
Inmunidad Innata/inmunología , Inflamación/inmunología , Piel/inmunología , Linfocitos T Reguladores/inmunología , Factor de Crecimiento Transformador beta/inmunología , Animales , Ratones , Ratones Congénicos , Ratones Endogámicos C57BL , Ratones Transgénicos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA