Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
1.
Am J Respir Crit Care Med ; 207(9): 1171-1182, 2023 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-36796082

RESUMEN

Rationale: Remodeling and loss of distal conducting airways, including preterminal and terminal bronchioles (pre-TBs/TBs), underlie progressive airflow limitation in chronic obstructive pulmonary disease (COPD). The cellular basis of these structural changes remains unknown. Objectives: To identify biological changes in pre-TBs/TBs in COPD at single-cell resolution and determine their cellular origin. Methods: We established a novel method of distal airway dissection and performed single-cell transcriptomic profiling of 111,412 cells isolated from different airway regions of 12 healthy lung donors and pre-TBs of 5 patients with COPD. Imaging CyTOF and immunofluorescence analysis of pre-TBs/TBs from 24 healthy lung donors and 11 subjects with COPD were performed to characterize cellular phenotypes at a tissue level. Region-specific differentiation of basal cells isolated from proximal and distal airways was studied using an air-liquid interface model. Measurements and Main Results: The atlas of cellular heterogeneity along the proximal-distal axis of the human lung was assembled and identified region-specific cellular states, including SCGB3A2+ SFTPB+ terminal airway-enriched secretory cells (TASCs) unique to distal airways. TASCs were lost in COPD pre-TBs/TBs, paralleled by loss of region-specific endothelial capillary cells, increased frequency of CD8+ T cells normally enriched in proximal airways, and augmented IFN-γ signaling. Basal cells residing in pre-TBs/TBs were identified as a cellular origin of TASCs. Regeneration of TASCs by these progenitors was suppressed by IFN-γ. Conclusions: Altered maintenance of the unique cellular organization of pre-TBs/TBs, including loss of the region-specific epithelial differentiation in these bronchioles, represents the cellular manifestation and likely the cellular basis of distal airway remodeling in COPD.


Asunto(s)
Linfocitos T CD8-positivos , Enfermedad Pulmonar Obstructiva Crónica , Humanos , Pulmón , Bronquiolos , Diagnóstico por Imagen
3.
Nat Methods ; 19(12): 1653-1661, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36316562

RESUMEN

Multiplexed imaging and spatial transcriptomics enable highly resolved spatial characterization of cellular phenotypes, but still largely depend on laborious manual annotation to understand higher-order patterns of tissue organization. As a result, higher-order patterns of tissue organization are poorly understood and not systematically connected to disease pathology or clinical outcomes. To address this gap, we developed an approach called UTAG to identify and quantify microanatomical tissue structures in multiplexed images without human intervention. Our method combines information on cellular phenotypes with the physical proximity of cells to accurately identify organ-specific microanatomical domains in healthy and diseased tissue. We apply our method to various types of images across healthy and disease states to show that it can consistently detect higher-level architectures in human tissues, quantify structural differences between healthy and diseased tissue, and reveal tissue organization patterns at the organ scale.


Asunto(s)
Diagnóstico por Imagen , Transcriptoma , Humanos
11.
Am J Respir Crit Care Med ; 196(3): 340-352, 2017 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-28345955

RESUMEN

RATIONALE: Small airways are the primary site of pathologic changes in chronic obstructive pulmonary disease (COPD), the major smoking-induced lung disorder. OBJECTIVES: On the basis of the concept of proximal-distal patterning that determines regional specialization of the airway epithelium during lung development, we hypothesized that a similar program operates in the adult human lung being altered by smoking, leading to decreased regional identity of the small airway epithelium (SAE). METHODS: The proximal and distal airway signatures were identified by comparing the transcriptomes of large and small airway epithelium samples obtained by bronchoscopy from healthy nonsmokers. The expression of these signatures was evaluated in the SAE of healthy smokers and smokers with COPD compared with that of healthy nonsmokers. The capacity of airway basal stem cells (BCs) to maintain region-associated phenotypes was evaluated using the air-liquid interface model. MEASUREMENTS AND MAIN RESULTS: The distal and proximal airway signatures, containing 134 and 233 genes, respectively, were identified. These signatures included known developmental regulators of airway patterning, as well as novel regulators such as epidermal growth factor receptor, which was associated with the proximal airway phenotype. In the SAE of smokers with COPD, there was a dramatic smoking-dependent loss of the regional transcriptome identity with concomitant proximalization. This repatterning phenotype was reproduced by stimulating SAE BCs with epidermal growth factor, which was up-regulated in the SAE of smokers, during differentiation of SAE BCs in vitro. CONCLUSIONS: Smoking-induced global distal-to-proximal reprogramming of the SAE represents a novel pathologic feature of COPD and is mediated by exaggerated epidermal growth factor/epidermal growth factor receptor signaling in SAE BCs.


Asunto(s)
Pulmón/fisiopatología , Enfermedad Pulmonar Obstructiva Crónica/fisiopatología , Fumar/fisiopatología , Adulto , Epitelio/fisiopatología , Femenino , Humanos , Masculino
12.
Stem Cells ; 35(3): 824-837, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-27709733

RESUMEN

The airway epithelium of cigarette smokers undergoes dramatic remodeling with hyperplasia of basal cells (BC) and mucus-producing cells, squamous metaplasia, altered ciliated cell differentiation and decreased junctional barrier integrity, relevant to chronic obstructive pulmonary disease and lung cancer. In this study, we show that epidermal growth factor receptor (EGFR) ligand amphiregulin (AREG) is induced by smoking in human airway epithelium as a result of epidermal growth factor (EGF)-driven squamous differentiation of airway BC stem/progenitor cells. In turn, AREG induced a unique EGFR activation pattern in human airway BC, distinct from that evoked by EGF, leading to BC- and mucous hyperplasia, altered ciliated cell differentiation and impaired barrier integrity. Further, AREG promoted its own expression and suppressed expression of EGF, establishing an autonomous self-amplifying signaling loop in airway BC relevant for promotion of EGF-independent hyperplastic phenotypes. Thus, EGF-AREG interplay in airway BC stem/progenitor cells is one of the mechanisms that mediates the interconnected pathogenesis of all major smoking-induced lesions in the human airway epithelium. Stem Cells 2017;35:824-837.


Asunto(s)
Anfirregulina/metabolismo , Factor de Crecimiento Epidérmico/metabolismo , Mucosa Respiratoria/patología , Fumar/efectos adversos , Células Madre/patología , Adulto , Remodelación de las Vías Aéreas (Respiratorias) , Diferenciación Celular , Proliferación Celular , Cilios/metabolismo , Regulación hacia Abajo , Receptores ErbB/metabolismo , Femenino , Humanos , Hiperplasia , Masculino , Células Madre/metabolismo , Regulación hacia Arriba
13.
Am J Respir Crit Care Med ; 194(5): 587-95, 2016 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-27007171

RESUMEN

RATIONALE: Waterpipes, also called hookahs, are currently used by millions of people worldwide. Despite the increasing use of waterpipe smoking, there is limited data on the health effects of waterpipe smoking and there are no federal regulations regarding its use. OBJECTIVES: To assess the effects of waterpipe smoking on the human lung using clinical and biological parameters in young, light-use waterpipe smokers. METHODS: We assessed young, light-use, waterpipe-only smokers in comparison with lifelong nonsmokers using clinical parameters of cough and sputum scores, lung function, and chest high-resolution computed tomography as well as biological parameters of lung epithelial lining fluid metabolome, small airway epithelial (SAE) cell differential and transcriptome, alveolar macrophage transcriptome, and plasma apoptotic endothelial cell microparticles. MEASUREMENTS AND MAIN RESULTS: Compared with nonsmokers, waterpipe smokers had more cough and sputum as well as a lower lung diffusing capacity, abnormal epithelial lining fluid metabolome profile, increased proportions of SAE secretory and intermediate cells, reduced proportions of SAE ciliated and basal cells, markedly abnormal SAE and alveolar macrophage transcriptomes, and elevated levels of apoptotic endothelial cell microparticles. CONCLUSIONS: Young, light-use, waterpipe-only smokers have a variety of abnormalities in multiple lung-related biological and clinical parameters, suggesting that even limited waterpipe use has broad consequences on human lung biology and health. We suggest that large epidemiological studies should be initiated to investigate the harmful effects of waterpipe smoking.


Asunto(s)
Pulmón/patología , Pulmón/fisiopatología , Capacidad de Difusión Pulmonar , Fumar/efectos adversos , Tabaquismo/complicaciones , Transcriptoma/efectos de los fármacos , Adulto , Monóxido de Carbono/análisis , Carboxihemoglobina/análisis , Estudios de Casos y Controles , Micropartículas Derivadas de Células/efectos de los fármacos , Cotinina/orina , Tos/etiología , Tos/microbiología , Células Epiteliales/efectos de los fármacos , Femenino , Volumen Espiratorio Forzado/fisiología , Humanos , Masculino , Nicotina/orina , Alveolos Pulmonares/citología , Alveolos Pulmonares/efectos de los fármacos , Esputo/química , Esputo/efectos de los fármacos , Tórax/diagnóstico por imagen , Tomografía Computarizada por Rayos X , Adulto Joven
14.
J Immunol ; 196(7): 3159-67, 2016 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-26927796

RESUMEN

In the process of seeking novel lung host defense regulators by analyzing genome-wide RNA sequence data from normal human airway epithelium, we detected expression of POU domain class 2-associating factor 1 (POU2AF1), a known transcription cofactor previously thought to be expressed only in lymphocytes. Lymphocyte contamination of human airway epithelial samples obtained by bronchoscopy and brushing was excluded by immunohistochemistry staining, the observation of upregulation of POU2AF1 in purified airway basal stem/progenitor cells undergoing differentiation, and analysis of differentiating single basal cell clones. Lentivirus-mediated upregulation of POU2AF1 in airway basal cells induced upregulation of host defense genes, including MX1, IFIT3, IFITM, and known POU2AF1 downstream genes HLA-DRA, ID2, ID3, IL6, and BCL6. Interestingly, expression of these genes paralleled changes of POU2AF1 expression during airway epithelium differentiation in vitro, suggesting POU2AF1 helps to maintain a host defense tone even in pathogen-free condition. Cigarette smoke, a known risk factor for airway infection, suppressed POU2AF1 expression both in vivo in humans and in vitro in human airway epithelial cultures, accompanied by deregulation of POU2AF1 downstream genes. Finally, enhancing POU2AF1 expression in human airway epithelium attenuated the suppression of host defense genes by smoking. Together, these findings suggest a novel function of POU2AF1 as a potential regulator of host defense genes in the human airway epithelium.


Asunto(s)
Regulación de la Expresión Génica , Inmunidad/genética , Mucosa Respiratoria/inmunología , Mucosa Respiratoria/metabolismo , Transactivadores/genética , Transactivadores/metabolismo , Diferenciación Celular , Análisis por Conglomerados , Células Epiteliales/metabolismo , Perfilación de la Expresión Génica , Humanos , Mucosa Respiratoria/citología , Fumar/efectos adversos
16.
Annu Rev Physiol ; 77: 379-406, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25386990

RESUMEN

A characteristic feature of the human airway epithelium is the presence of ciliated cells bearing motile cilia, specialized cell surface projections containing axonemes composed of microtubules and dynein arms, which provide ATP-driven motility. In the airways, cilia function in concert with airway mucus to mediate the critical function of mucociliary clearance, cleansing the airways of inhaled particles and pathogens. The prototypical disorder of respiratory cilia is primary ciliary dyskinesia, an inherited disorder that leads to impaired mucociliary clearance, to repeated chest infections, and to the progressive destruction of lung architecture. Numerous acquired lung diseases are also marked by abnormalities in both cilia structure and function. In this review we summarize current knowledge regarding airway ciliated cells and cilia, how they function to maintain a healthy epithelium, and how disorders of cilia structure and function contribute to inherited and acquired lung disease.


Asunto(s)
Cilios/fisiología , Trastornos de la Motilidad Ciliar/fisiopatología , Enfermedades Pulmonares/fisiopatología , Mucosa Respiratoria/fisiopatología , Animales , Axonema/fisiología , Modelos Animales de Enfermedad , Humanos , Ratones , Microtúbulos/fisiología , Depuración Mucociliar/fisiología
17.
Ann Am Thorac Soc ; 11 Suppl 5: S252-8, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25525728

RESUMEN

The airway epithelium is the primary site of the earliest pathologic changes induced by smoking, contributing to the development of chronic obstructive pulmonary disease (COPD). The normal human airway epithelium is composed of several major cell types, including differentiated ciliated and secretory cells, intermediate undifferentiated cells, and basal cells (BC). BC contain the stem/progenitor cell population responsible for maintenance of the normally differentiated airway epithelium. Although inflammatory and immune processes play a significant role in the pathogenesis of COPD, the earliest lesions include hyperplasia of the BC population, suggesting that the disease may start with this cell type. Apart from BC hyperplasia, smoking induces a number of COPD-relevant airway epithelial remodeling phenotypes that are likely initiated in the BC population, including mucous cell hyperplasia, squamous cell metaplasia, epithelial-mesenchymal transition, altered ciliated and nonmucous secretory cell differentiation, and suppression of junctional barrier integrity. Significant progress has been recently made in understanding the biology of human airway BC, including gene expression features, stem/progenitor, and other functions, including interaction with other airway cell types. Accumulating evidence suggests that human airway BC function as both sensors and cellular sources of various cytokines and growth factors relevant to smoking-associated airway injury, as well as the origin of various molecular and histological phenotypes relevant to the pathogenesis of COPD. In the context of these considerations, we suggest that early BC-specific smoking-induced molecular changes are critical to the pathogenesis of COPD, and these represent a candidate target for novel therapeutic approaches to prevent COPD progression in susceptible individuals.


Asunto(s)
Citocinas/genética , ADN/genética , Regulación de la Expresión Génica , Enfermedad Pulmonar Obstructiva Crónica/genética , Sistema Respiratorio/patología , Fumar/efectos adversos , Células Madre/fisiología , Diferenciación Celular , Citocinas/biosíntesis , Humanos , Fenotipo , Enfermedad Pulmonar Obstructiva Crónica/metabolismo , Enfermedad Pulmonar Obstructiva Crónica/patología , Mucosa Respiratoria , Sistema Respiratorio/metabolismo , Factores de Tiempo
19.
PLoS One ; 9(2): e88051, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24498427

RESUMEN

Genome-wide association studies (GWAS) and candidate gene studies have identified a number of risk loci associated with the smoking-related disease COPD, a disorder that originates in the airway epithelium. Since airway basal cell (BC) stem/progenitor cells exhibit the earliest abnormalities associated with smoking (hyperplasia, squamous metaplasia), we hypothesized that smoker BC have a dysregulated transcriptome, enriched, in part, at known GWAS/candidate gene loci. Massive parallel RNA sequencing was used to compare the transcriptome of BC purified from the airway epithelium of healthy nonsmokers (n = 10) and healthy smokers (n = 7). The chromosomal location of the differentially expressed genes was compared to loci identified by GWAS to confer risk for COPD. Smoker BC have 676 genes differentially expressed compared to nonsmoker BC, dominated by smoking up-regulation. Strikingly, 166 (25%) of these genes are located on chromosome 19, with 13 localized to 19q13.2 (p<10⁻4 compared to chance), including 4 genes (NFKBIB, LTBP4, EGLN2 and TGFB1) associated with risk for COPD. These observations provide the first direct connection between known genetic risks for smoking-related lung disease and airway BC, the population of lung cells that undergo the earliest changes associated with smoking.


Asunto(s)
Biomarcadores/metabolismo , Cromosomas Humanos Par 19/genética , Células Endoteliales/metabolismo , Perfilación de la Expresión Génica , Sitios Genéticos , Enfermedad Pulmonar Obstructiva Crónica/genética , Mucosa Respiratoria/metabolismo , Fumar/efectos adversos , Adulto , Variaciones en el Número de Copia de ADN/genética , Metilación de ADN , Estudio de Asociación del Genoma Completo , Humanos , Análisis de Secuencia por Matrices de Oligonucleótidos , Enfermedad Pulmonar Obstructiva Crónica/inducido químicamente , Enfermedad Pulmonar Obstructiva Crónica/patología , Mucosa Respiratoria/efectos de los fármacos , Mucosa Respiratoria/patología , Fumar/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...