Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
1.
Pathogens ; 13(4)2024 Mar 29.
Artículo en Inglés | MEDLINE | ID: mdl-38668247

RESUMEN

The transient depletion of monocytes alone prior to exposure of macaques to HTLV-1 enhances both HTLV-1WT (wild type) and HTLV-1p12KO (Orf-1 knockout) infectivity, but seroconversion to either virus is not sustained over time, suggesting a progressive decrease in virus expression. These results raise the hypotheses that either HTLV-1 persistence depends on a monocyte reservoir or monocyte depletion provides a transient immune evasion benefit. To test these hypotheses, we simultaneously depleted NK cells, CD8+ T cells, and monocytes (triple depletion) prior to exposure to HTLV-1WT or HTLV-1p12KO. Remarkably, triple depletion resulted in exacerbation of infection by both viruses and complete rescue of HTLV-1p12KO infectivity. Following triple depletion, we observed rapid and sustained seroconversion, high titers of antibodies against HTLV-1 p24Gag, and frequent detection of viral DNA in the blood and tissues of all animals when compared with depletion of only CD8+ and NK cells, or monocytes alone. The infection of macaques with HTLV-1WT or HTLV-1p12KO was associated with higher plasma levels of IL-10 after 21 weeks, while IL-6, IFN-γ, IL-18, and IL-1ß were only elevated in animals infected with HTLV-1WT. The repeat depletion of monocytes, NK, and CD8+ cells seven months following the first exposure to HTLV-1 did not further exacerbate viral replication. These results underscore the contribution of monocytes in orchestrating anti-viral immunity. Indeed, the absence of orf-1 expression was fully compensated by the simultaneous depletion of CD8+ T cells, NK cells, and monocytes, underlining the primary role of orf-1 in hijacking host immunity.

2.
Cell Rep ; 42(5): 112501, 2023 05 30.
Artículo en Inglés | MEDLINE | ID: mdl-37178117

RESUMEN

Locoregional monotherapy with heterodimeric interleukin (IL)-15 (hetIL-15) in a triple-negative breast cancer (TNBC) orthotopic mouse model resulted in tumor eradication in 40% of treated mice, reduction of metastasis, and induction of immunological memory against breast cancer cells. hetIL-15 re-shaped the tumor microenvironment by promoting the intratumoral accumulation of cytotoxic lymphocytes, conventional type 1 dendritic cells (cDC1s), and a dendritic cell (DC) population expressing both CD103 and CD11b markers. These CD103intCD11b+DCs share phenotypic and gene expression characteristics with both cDC1s and cDC2s, have transcriptomic profiles more similar to monocyte-derived DCs (moDCs), and correlate with tumor regression. Therefore, hetIL-15, a cytokine directly affecting lymphocytes and inducing cytotoxic cells, also has an indirect rapid and significant effect on the recruitment of myeloid cells, initiating a cascade for tumor elimination through innate and adoptive immune mechanisms. The intratumoral CD103intCD11b+DC population induced by hetIL-15 may be targeted for the development of additional cancer immunotherapy approaches.


Asunto(s)
Antineoplásicos , Neoplasias , Ratones , Animales , Cadenas alfa de Integrinas/metabolismo , Neoplasias/metabolismo , Citocinas/metabolismo , Células Dendríticas/metabolismo , Linfocitos/metabolismo , Antineoplásicos/metabolismo , Factores Inmunológicos/metabolismo , Ratones Endogámicos C57BL , Microambiente Tumoral
3.
Cell Rep ; 32(1): 107872, 2020 07 07.
Artículo en Inglés | MEDLINE | ID: mdl-32640237

RESUMEN

The type 2 cytokine-high asthma endotype (T2H) is characterized by IL-13-driven mucus obstruction of the airways. To further investigate this incompletely understood pathobiology, we characterize IL-13 effects on human airway epithelial cell cultures using single-cell RNA sequencing, finding that IL-13 generates a distinctive transcriptional state for each cell type. Specifically, we discover a mucus secretory program induced by IL-13 in all cell types which converts both mucus and defense secretory cells into a metaplastic state with emergent mucin production and secretion, while leading to ER stress and cell death in ciliated cells. The IL-13-remodeled epithelium secretes a pathologic, mucin-imbalanced, and innate immunity-depleted proteome that arrests mucociliary motion. Signatures of IL-13-induced cellular remodeling are mirrored by transcriptional signatures characteristic of the nasal airway epithelium within T2H versus T2-low asthmatic children. Our results reveal the epithelium-wide scope of T2H asthma and present candidate therapeutic targets for restoring normal epithelial function.


Asunto(s)
Asma/genética , Epitelio/metabolismo , Análisis de la Célula Individual , Transcriptoma/genética , Transporte Biológico/efectos de los fármacos , Reprogramación Celular/efectos de los fármacos , Niño , Cilios/efectos de los fármacos , Cilios/metabolismo , Regulación hacia Abajo/efectos de los fármacos , Estrés del Retículo Endoplásmico/efectos de los fármacos , Epitelio/efectos de los fármacos , Humanos , Inmunidad Innata/efectos de los fármacos , Interferones/metabolismo , Interleucina-13/farmacología , Metaplasia , Moco/metabolismo , Transcriptoma/efectos de los fármacos
4.
Nat Commun ; 11(1): 2485, 2020 05 19.
Artículo en Inglés | MEDLINE | ID: mdl-32427931

RESUMEN

Cigarette smoke first interacts with the lung through the cellularly diverse airway epithelium and goes on to drive development of most chronic lung diseases. Here, through single cell RNA-sequencing analysis of the tracheal epithelium from smokers and non-smokers, we generate a comprehensive atlas of epithelial cell types and states, connect these into lineages, and define cell-specific responses to smoking. Our analysis infers multi-state lineages that develop into surface mucus secretory and ciliated cells and then contrasts these to the unique specification of submucosal gland (SMG) cells. Accompanying knockout studies reveal that tuft-like cells are the likely progenitor of both pulmonary neuroendocrine cells and CFTR-rich ionocytes. Our smoking analysis finds that all cell types, including protected stem and SMG populations, are affected by smoking through both pan-epithelial smoking response networks and hundreds of cell-specific response genes, redefining the penetrance and cellular specificity of smoking effects on the human airway epithelium.


Asunto(s)
Células Epiteliales/metabolismo , Perfilación de la Expresión Génica/métodos , Pulmón/metabolismo , Mucosa Respiratoria/metabolismo , Fumar/genética , Tráquea/metabolismo , Animales , Células Cultivadas , Técnicas de Inactivación de Genes , Redes Reguladoras de Genes , Humanos , Pulmón/citología , Ratones , Células 3T3 NIH , No Fumadores/estadística & datos numéricos , Mucosa Respiratoria/citología , Análisis de Secuencia de ARN/métodos , Análisis de la Célula Individual/métodos , Fumadores/estadística & datos numéricos , Tráquea/citología
5.
Am J Respir Cell Mol Biol ; 63(2): 172-184, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32275839

RESUMEN

Air pollution particulate matter <2.5 µm (PM2.5) exposure is associated with poor respiratory outcomes. Mechanisms underlying PM2.5-induced lung pathobiology are poorly understood but likely involve cellular and molecular changes to the airway epithelium. We extracted and chemically characterized the organic and water-soluble components of air pollution PM2.5 samples, then determined the whole transcriptome response of human nasal mucociliary airway epithelial cultures to a dose series of PM2.5 extracts. We found that PM2.5 organic extract (OE), but not water-soluble extract, elicited a potent, dose-dependent transcriptomic response from the mucociliary epithelium. Exposure to a moderate OE dose modified the expression of 424 genes, including activation of aryl hydrocarbon receptor signaling and an IL-1 inflammatory program. We generated an OE-response gene network defined by eight functional enrichment groups, which exhibited high connectivity through CYP1A1, IL1A, and IL1B. This OE exposure also robustly activated a mucus secretory expression program (>100 genes), which included transcriptional drivers of mucus metaplasia (SPDEF and FOXA3). Exposure to a higher OE dose modified the expression of 1,240 genes and further exacerbated expression responses observed at the moderate dose, including the mucus secretory program. Moreover, the higher OE dose significantly increased the MUC5AC/MUC5B gel-forming mucin expression ratio and strongly downregulated ciliated cell expression programs, including key ciliating cell transcription factors (e.g., FOXJ1 and MCIDAS). Chronic OE stimulation induced mucus metaplasia-like remodeling characterized by increases in MUC5AC+ secretory cells and MUC5AC mucus secretions. This epithelial remodeling may underlie poor respiratory outcomes associated with high PM2.5 exposure.


Asunto(s)
Mucosa Nasal/diagnóstico por imagen , Material Particulado/efectos adversos , Mucosa Respiratoria/efectos de los fármacos , Contaminantes Atmosféricos/efectos adversos , Contaminación del Aire/efectos adversos , Asma/inducido químicamente , Asma/genética , Estudio de Asociación del Genoma Completo/métodos , Humanos , Inflamación/inducido químicamente , Inflamación/genética , Mucina 5AC/genética , Mucina 5B/genética , Factores de Transcripción/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA