Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
J Exp Med ; 220(12)2023 12 04.
Artículo en Inglés | MEDLINE | ID: mdl-37812390

RESUMEN

Cutaneous leishmaniasis causes alterations in the skin microbiota, leading to pathologic immune responses and delayed healing. However, it is not known how these microbiota-driven immune responses are regulated. Here, we report that depletion of Foxp3+ regulatory T cells (Tregs) in Staphylococcus aureus-colonized mice resulted in less IL-17 and an IFN-γ-dependent skin inflammation with impaired S. aureus immunity. Similarly, reducing Tregs in S. aureus-colonized and Leishmania braziliensis-infected mice increased IFN-γ, S. aureus, and disease severity. Importantly, analysis of lesions from L. braziliensis patients revealed that low FOXP3 gene expression is associated with high IFNG expression, S. aureus burden, and delayed lesion resolution compared to patients with high FOXP3 expression. Thus, we found a critical role for Tregs in regulating the balance between IL-17 and IFN-γ in the skin, which influences both bacterial burden and disease. These results have clinical ramifications for cutaneous leishmaniasis and other skin diseases associated with a dysregulated microbiome when Tregs are limited or dysfunctional.


Asunto(s)
Leishmaniasis Cutánea , Infecciones Estafilocócicas , Humanos , Animales , Ratones , Staphylococcus aureus , Interleucina-17 , Linfocitos T Reguladores , Gravedad del Paciente , Factores de Transcripción Forkhead
2.
Cell Rep ; 42(10): 113281, 2023 10 31.
Artículo en Inglés | MEDLINE | ID: mdl-37858460

RESUMEN

Strain-level variation in Staphylococcus aureus is a factor that contributes to disease burden and clinical outcomes in skin disorders and chronic wounds. However, the microbial mechanisms that drive these variable host responses are poorly understood. To identify mechanisms underlying strain-specific outcomes, we perform high-throughput phenotyping screens on S. aureus isolates cultured from diabetic foot ulcers. Isolates from non-healing wounds produce more staphyloxanthin, a cell membrane pigment. In murine diabetic wounds, staphyloxanthin-producing isolates delay wound closure significantly compared with staphyloxanthin-deficient isolates. Staphyloxanthin promotes resistance to oxidative stress and enhances bacterial survival in neutrophils. Comparative genomic and transcriptomic analysis of genetically similar clinical isolates with disparate staphyloxanthin phenotypes reveals a mutation in the sigma B operon, resulting in marked differences in stress response gene expression. Our work illustrates a framework to identify traits that underlie strain-level variation in disease burden and suggests more precise targets for therapeutic intervention in S. aureus-positive wounds.


Asunto(s)
Diabetes Mellitus , Infecciones Estafilocócicas , Animales , Ratones , Staphylococcus aureus/metabolismo , Infecciones Estafilocócicas/microbiología , Cicatrización de Heridas
3.
Sci Transl Med ; 15(718): eadh1469, 2023 10 18.
Artículo en Inglés | MEDLINE | ID: mdl-37851822

RESUMEN

Leishmania braziliensis is a parasitic infection that can result in inflammation and skin injury with highly variable and unpredictable clinical outcomes. Here, we investigated the potential impact of microbiota on infection-induced inflammatory responses and disease resolution by conducting an integrated analysis of the skin microbiome and host transcriptome on a cohort of 62 patients infected with L. braziliensis. We found that overall bacterial burden and microbiome configurations dominated with Staphylococcus spp. were associated with delayed healing and enhanced inflammatory responses, especially by IL-1 family members. Quantification of host and bacterial transcripts on human lesions revealed that high lesional S. aureus transcript abundance was associated with delayed healing and increased expression of IL-1ß. This cytokine was critical for modulating disease outcomes in L. braziliensis-infected mice colonized with S. aureus, given that its neutralization reduced pathology and inflammation. These results highlight how the human microbiome can shape disease outcomes in cutaneous leishmaniasis and suggest pathways toward host-directed therapies to mitigate the inflammatory consequences.


Asunto(s)
Leishmaniasis Cutánea , Microbiota , Humanos , Ratones , Animales , Staphylococcus aureus , Multiómica , Inflamación , Bacterias , Gravedad del Paciente
4.
medRxiv ; 2023 Feb 06.
Artículo en Inglés | MEDLINE | ID: mdl-36798406

RESUMEN

Leishmania braziliensis infection results in inflammation and skin injury, with highly variable and unpredictable clinical outcomes. Here, we investigated the potential impact of microbiota on infection-induced inflammatory responses and disease resolution by conducting an integrated analysis of the skin microbiome and host transcriptome on a cohort of 62 L. braziliensis -infected patients. We found that overall bacterial burden and microbiome configurations dominated with Staphylococcus spp. were associated with delayed healing and enhanced inflammatory responses, especially by IL-1 family members. Dual RNA-seq of human lesions revealed that high lesional S. aureus transcript abundance was associated with delayed healing and increased expression of IL-1ß. This cytokine was critical for modulating disease outcome in L. braziliensis -infected mice colonized with S. aureus , as its neutralization reduced pathology and inflammation. These results implicate the microbiome in cutaneous leishmaniasis disease outcomes in humans and suggest host-directed therapies to mitigate the inflammatory consequences.

5.
Cell Host Microbe ; 29(8): 1235-1248.e8, 2021 Aug 11.
Artículo en Inglés | MEDLINE | ID: mdl-34214492

RESUMEN

The epidermis forms a barrier that defends the body from desiccation and entry of harmful substances, while also sensing and integrating environmental signals. The tightly orchestrated cellular changes needed for the formation and maintenance of this epidermal barrier occur in the context of the skin microbiome. Using germ-free mice, we demonstrate the microbiota is necessary for proper differentiation and repair of the epidermal barrier. These effects are mediated by microbiota signaling through the aryl hydrocarbon receptor (AHR) in keratinocytes, a xenobiotic receptor also implicated in epidermal differentiation. Mice lacking keratinocyte AHR are more susceptible to barrier damage and infection, during steady-state and epicutaneous sensitization. Colonization with a defined consortium of human skin isolates restored barrier competence in an AHR-dependent manner. We reveal a fundamental mechanism whereby the microbiota regulates skin barrier formation and repair, which has far-reaching implications for the numerous skin disorders characterized by epidermal barrier dysfunction.


Asunto(s)
Microbiota/fisiología , Receptores de Hidrocarburo de Aril/metabolismo , Transducción de Señal , Piel/microbiología , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico , Diferenciación Celular , Línea Celular , Células Epidérmicas/metabolismo , Células Epidérmicas/patología , Epidermis/metabolismo , Femenino , Humanos , Queratinocitos , Masculino , Ratones , Ratones Endogámicos C57BL , Piel/patología , Enfermedades de la Piel/microbiología
6.
Proc Natl Acad Sci U S A ; 117(2): 1160-1166, 2020 01 14.
Artículo en Inglés | MEDLINE | ID: mdl-31879349

RESUMEN

Following mycobacterial entry into macrophages the ESX-1 type VII secretion system promotes phagosomal permeabilization and type I IFN production, key features of tuberculosis pathogenesis. The current model states that the secreted substrate ESAT-6 is required for membrane permeabilization and that a subsequent passive leakage of extracellular bacterial DNA into the host cell cytosol is sensed by the cyclic GMP-AMP synthase (cGAS) and stimulator of IFN genes (STING) pathway to induce type I IFN production. We employed a collection of Mycobacterium marinum ESX-1 transposon mutants in a macrophage infection model and show that permeabilization of the phagosomal membrane does not require ESAT-6 secretion. Moreover, loss of membrane integrity is insufficient to induce type I IFN production. Instead, type I IFN production requires intact ESX-1 function and correlates with release of mitochondrial and nuclear host DNA into the cytosol, indicating that ESX-1 affects host membrane integrity and DNA release via genetically separable mechanisms. These results suggest a revised model for major aspects of ESX-1-mediated host interactions and put focus on elucidating the mechanisms by which ESX-1 permeabilizes host membranes and induces the type I IFN response, questions of importance for our basic understanding of mycobacterial pathogenesis and innate immune sensing.


Asunto(s)
Antígenos Bacterianos/metabolismo , Permeabilidad de la Membrana Celular/fisiología , Interferón Tipo I/metabolismo , Infecciones por Mycobacterium no Tuberculosas/metabolismo , Mycobacterium marinum/patogenicidad , Fagosomas/metabolismo , Antígenos Bacterianos/genética , Proteínas Bacterianas/metabolismo , Interacciones Huésped-Patógeno/inmunología , Macrófagos/metabolismo , Macrófagos/microbiología , Mitocondrias/metabolismo , Infecciones por Mycobacterium no Tuberculosas/microbiología , Mycobacterium marinum/genética , Mycobacterium marinum/inmunología , Mycobacterium marinum/metabolismo , Tuberculosis/inmunología , Sistemas de Secreción Tipo VII
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA