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











Intervalo de año de publicación
1.
Nat Commun ; 15(1): 3900, 2024 May 09.
Artículo en Inglés | MEDLINE | ID: mdl-38724552

RESUMEN

By incompletely understood mechanisms, type 2 (T2) inflammation present in the airways of severe asthmatics drives the formation of pathologic mucus which leads to airway mucus plugging. Here we investigate the molecular role and clinical significance of intelectin-1 (ITLN-1) in the development of pathologic airway mucus in asthma. Through analyses of human airway epithelial cells we find that ITLN1 gene expression is highly induced by interleukin-13 (IL-13) in a subset of metaplastic MUC5AC+ mucus secretory cells, and that ITLN-1 protein is a secreted component of IL-13-induced mucus. Additionally, we find ITLN-1 protein binds the C-terminus of the MUC5AC mucin and that its deletion in airway epithelial cells partially reverses IL-13-induced mucostasis. Through analysis of nasal airway epithelial brushings, we find that ITLN1 is highly expressed in T2-high asthmatics, when compared to T2-low children. Furthermore, we demonstrate that both ITLN-1 gene expression and protein levels are significantly reduced by a common genetic variant that is associated with protection from the formation of mucus plugs in T2-high asthma. This work identifies an important biomarker and targetable pathways for the treatment of mucus obstruction in asthma.


Asunto(s)
Asma , Proteínas Ligadas a GPI , Interleucina-13 , Lectinas , Mucina 5AC , Moco , Niño , Humanos , Asma/genética , Asma/metabolismo , Citocinas , Células Epiteliales/metabolismo , Proteínas Ligadas a GPI/genética , Proteínas Ligadas a GPI/metabolismo , Interleucina-13/genética , Interleucina-13/metabolismo , Lectinas/genética , Lectinas/metabolismo , Mucina 5AC/genética , Mucina 5AC/metabolismo , Moco/metabolismo , Mucosa Nasal/metabolismo , Polimorfismo Genético , Mucosa Respiratoria/metabolismo
2.
JCI Insight ; 7(13)2022 07 08.
Artículo en Inglés | MEDLINE | ID: mdl-35608904

RESUMEN

Chronic type 2 (T2) inflammatory diseases of the respiratory tract are characterized by mucus overproduction and disordered mucociliary function, which are largely attributed to the effects of IL-13 on common epithelial cell types (mucus secretory and ciliated cells). The role of rare cells in airway T2 inflammation is less clear, though tuft cells have been shown to be critical in the initiation of T2 immunity in the intestine. Using bulk and single-cell RNA sequencing of airway epithelium and mouse modeling, we found that IL-13 expanded and programmed airway tuft cells toward eicosanoid metabolism and that tuft cell deficiency led to a reduction in airway prostaglandin E2 (PGE2) concentration. Allergic airway epithelia bore a signature of PGE2 activation, and PGE2 activation led to cystic fibrosis transmembrane receptor-dependent ion and fluid secretion and accelerated mucociliary transport. These data reveal a role for tuft cells in regulating epithelial mucociliary function in the allergic airway.


Asunto(s)
Regulador de Conductancia de Transmembrana de Fibrosis Quística , Fibrosis Quística , Animales , Fibrosis Quística/genética , Regulador de Conductancia de Transmembrana de Fibrosis Quística/metabolismo , Dinoprostona , Interleucina-13/metabolismo , Ratones , Sistema Respiratorio
3.
Nat Commun ; 11(1): 5139, 2020 10 12.
Artículo en Inglés | MEDLINE | ID: mdl-33046696

RESUMEN

Coronavirus disease 2019 (COVID-19) is caused by SARS-CoV-2, an emerging virus that utilizes host proteins ACE2 and TMPRSS2 as entry factors. Understanding the factors affecting the pattern and levels of expression of these genes is important for deeper understanding of SARS-CoV-2 tropism and pathogenesis. Here we explore the role of genetics and co-expression networks in regulating these genes in the airway, through the analysis of nasal airway transcriptome data from 695 children. We identify expression quantitative trait loci for both ACE2 and TMPRSS2, that vary in frequency across world populations. We find TMPRSS2 is part of a mucus secretory network, highly upregulated by type 2 (T2) inflammation through the action of interleukin-13, and that the interferon response to respiratory viruses highly upregulates ACE2 expression. IL-13 and virus infection mediated effects on ACE2 expression were also observed at the protein level in the airway epithelium. Finally, we define airway responses to common coronavirus infections in children, finding that these infections generate host responses similar to other viral species, including upregulation of IL6 and ACE2. Our results reveal possible mechanisms influencing SARS-CoV-2 infectivity and COVID-19 clinical outcomes.


Asunto(s)
Betacoronavirus/fisiología , Infecciones por Coronavirus/virología , Interferones/metabolismo , Interleucina-13/metabolismo , Mucosa Nasal/patología , Peptidil-Dipeptidasa A/genética , Neumonía Viral/virología , Serina Endopeptidasas/genética , Enzima Convertidora de Angiotensina 2 , COVID-19 , Niño , Infecciones por Coronavirus/metabolismo , Infecciones por Coronavirus/patología , Células Epiteliales/metabolismo , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Variación Genética , Interacciones Huésped-Patógeno , Humanos , Inflamación , Persona de Mediana Edad , Mucosa Nasal/metabolismo , Pandemias , Peptidil-Dipeptidasa A/metabolismo , Neumonía Viral/metabolismo , Neumonía Viral/patología , SARS-CoV-2 , Serina Endopeptidasas/metabolismo , Internalización del Virus
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
6.
Environ Sci Technol ; 39(24): 9725-31, 2005 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-16475359

RESUMEN

RDX (hexahydro-1,3,5-trinitro-1,3,5-triazine) and HMX (octahydro-1,3,5,7-tetranitro-1,3,5,7-tetrazocine) are cyclic nitramines ((CH2NNO2)n; n = 3 or 4, respectively) widely used as energetic chemicals. Their extensive use led to wide environmental contamination. In contrast to RDX, HMX tends to accumulate in soils due to its unique recalcitrance. In the present study, we investigated the potential of zerovalent iron (ZVI) to transform HMX under anoxic conditions. HMX underwent a rapid transformation when added in well-mixed anoxic ZVI-H2O batch systems to ultimately produce formaldehyde (HCHO), ammonium (NH4+), hydrazine (NH2NH2), and nitrous oxide (N2O). Time course experiments showed that the mechanism of HMX transformation occurred through at least two initial reactions. One reaction involved the sequential reduction of N-NO2 groups to the five nitroso products (1NO-HMX, cis-2NO-HMX, trans-2NO-HMX, 3NO-HMX, and 4NO-HMX). Another implied ring cleavage from either HMX or 1NO-HMX as demonstrated by the observation of methylenedinitramine (NH(NO2)CH2NH(NO2)) and another intermediate that was tentatively identified as (NH(NO2)CH2N(NO)CH2NH-(NO2)) or its isomer (NH(NO)CH2N(NO2)CH2NH(NO2)). This is the first study that demonstrates transformation of HMX by ZVI to significant amounts of NH2NH2 and HCHO. Both toxic products seemed to persist under reductive conditions, thereby suggesting that the ultimate fate of these chemicals, particularly hydrazine, should be understood prior to using zerovalent iron to remediate cyclic nitramines.


Asunto(s)
Azocinas/metabolismo , Compuestos Heterocíclicos con 1 Anillo/metabolismo , Hierro/química , Aguas del Alcantarillado/microbiología , Microbiología del Suelo , Contaminantes del Suelo/metabolismo , Anaerobiosis , Compuestos de Anilina/metabolismo , Azocinas/toxicidad , Formaldehído/metabolismo , Compuestos Heterocíclicos con 1 Anillo/toxicidad , Hidrazinas/metabolismo , Hierro/farmacología , Isomerismo , Nitrobencenos/metabolismo , Óxido Nitroso/metabolismo , Compuestos de Amonio Cuaternario/metabolismo , Contaminantes del Suelo/toxicidad , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA