Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 49
Filtrar
1.
Med Clin (Barc) ; 2024 Mar 26.
Artículo en Inglés, Español | MEDLINE | ID: mdl-38538430

RESUMEN

INTRODUCTION AND OBJECTIVES: Smoking is associated with various health risks, including cancer, cardiovascular disease, and chronic obstructive pulmonary disease. In this retrospective cohort study, we aimed to determine whether smoking is harmful to the whole metabolic system. METHODS: We collected data from 340 randomly selected participants who were divided into three groups: smokers (n=137), non-smokers (n=134), and ex-smokers (n=69). We obtained information on participants' body mass index, waist circumference, indicators of glucose metabolism, lipid metabolism, bone metabolism, and uric acid from health screen data during the past three years. A cluster analysis was used to synthesize each participant's overall metabolic characteristics. RESULTS: According to the cluster analysis, the 340 participants were divided into three groups: excellent metabolizers (137, 40.3%), adverse metabolizers (32, 9.4%), and intermediate metabolizers (171, 50.3%). The Chi-squared test analysis shows that people with different smoking statuses have different metabolic patterns. Non-smokers had the highest proportion of excellent metabolizers (56%), and current smokers had the highest proportion of adverse metabolizers (15.3%). The proportion of adverse metabolizers (5.8%) in the ex-smoker group was clinically relevantly lower than that of current smokers. CONCLUSION: The statistically significant differences in the distribution of smokers into different metabolic clusters indicate that smoking has adverse effects on the whole metabolic system of the human body, which further increases the existing global burden of metabolic disorders.

2.
Nature ; 624(7991): 378-389, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-38092917

RESUMEN

Recent advances in single-cell technologies have led to the discovery of thousands of brain cell types; however, our understanding of the gene regulatory programs in these cell types is far from complete1-4. Here we report a comprehensive atlas of candidate cis-regulatory DNA elements (cCREs) in the adult mouse brain, generated by analysing chromatin accessibility in 2.3 million individual brain cells from 117 anatomical dissections. The atlas includes approximately 1 million cCREs and their chromatin accessibility across 1,482 distinct brain cell populations, adding over 446,000 cCREs to the most recent such annotation in the mouse genome. The mouse brain cCREs are moderately conserved in the human brain. The mouse-specific cCREs-specifically, those identified from a subset of cortical excitatory neurons-are strongly enriched for transposable elements, suggesting a potential role for transposable elements in the emergence of new regulatory programs and neuronal diversity. Finally, we infer the gene regulatory networks in over 260 subclasses of mouse brain cells and develop deep-learning models to predict the activities of gene regulatory elements in different brain cell types from the DNA sequence alone. Our results provide a resource for the analysis of cell-type-specific gene regulation programs in both mouse and human brains.


Asunto(s)
Encéfalo , Cromatina , Análisis de la Célula Individual , Animales , Humanos , Ratones , Encéfalo/citología , Encéfalo/metabolismo , Corteza Cerebral/citología , Cromatina/química , Cromatina/genética , Cromatina/metabolismo , Aprendizaje Profundo , Elementos Transponibles de ADN/genética , Redes Reguladoras de Genes/genética , Neuronas/metabolismo
3.
Am J Respir Crit Care Med ; 208(6): 709-725, 2023 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-37463497

RESUMEN

Rationale: Alveolar capillary dysplasia with misalignment of pulmonary veins (ACDMPV) is a lethal developmental disorder of lung morphogenesis caused by insufficiency of FOXF1 (forkhead box F1) transcription factor function. The cellular and transcriptional mechanisms by which FOXF1 deficiency disrupts human lung formation are unknown. Objectives: To identify cell types, gene networks, and cell-cell interactions underlying the pathogenesis of ACDMPV. Methods: We used single-nucleus RNA and assay for transposase-accessible chromatin sequencing, immunofluorescence confocal microscopy, and RNA in situ hybridization to identify cell types and molecular networks influenced by FOXF1 in ACDMPV lungs. Measurements and Main Results: Pathogenic single-nucleotide variants and copy-number variant deletions involving the FOXF1 gene locus in all subjects with ACDMPV (n = 6) were accompanied by marked changes in lung structure, including deficient alveolar development and a paucity of pulmonary microvasculature. Single-nucleus RNA and assay for transposase-accessible chromatin sequencing identified alterations in cell number and gene expression in endothelial cells (ECs), pericytes, fibroblasts, and epithelial cells in ACDMPV lungs. Distinct cell-autonomous roles for FOXF1 in capillary ECs and pericytes were identified. Pathogenic variants involving the FOXF1 gene locus disrupt gene expression in EC progenitors, inhibiting the differentiation or survival of capillary 2 ECs and cell-cell interactions necessary for both pulmonary vasculogenesis and alveolar type 1 cell differentiation. Loss of the pulmonary microvasculature was associated with increased VEGFA (vascular endothelial growth factor A) signaling and marked expansion of systemic bronchial ECs expressing COL15A1 (collagen type XV α 1 chain). Conclusions: Distinct FOXF1 gene regulatory networks were identified in subsets of pulmonary endothelial and fibroblast progenitors, providing both cellular and molecular targets for the development of therapies for ACDMPV and other diffuse lung diseases of infancy.


Asunto(s)
Síndrome de Circulación Fetal Persistente , Recién Nacido , Humanos , Síndrome de Circulación Fetal Persistente/genética , Síndrome de Circulación Fetal Persistente/patología , Redes Reguladoras de Genes/genética , Factor A de Crecimiento Endotelial Vascular/genética , Células Endoteliales/patología , Multiómica , Pulmón/patología , ARN , Factores de Transcripción Forkhead/genética
4.
Proc Natl Acad Sci U S A ; 120(20): e2210991120, 2023 05 16.
Artículo en Inglés | MEDLINE | ID: mdl-37155843

RESUMEN

In 2021, the World Health Organization reclassified glioblastoma, the most common form of adult brain cancer, into isocitrate dehydrogenase (IDH)-wild-type glioblastomas and grade IV IDH mutant (G4 IDHm) astrocytomas. For both tumor types, intratumoral heterogeneity is a key contributor to therapeutic failure. To better define this heterogeneity, genome-wide chromatin accessibility and transcription profiles of clinical samples of glioblastomas and G4 IDHm astrocytomas were analyzed at single-cell resolution. These profiles afforded resolution of intratumoral genetic heterogeneity, including delineation of cell-to-cell variations in distinct cell states, focal gene amplifications, as well as extrachromosomal circular DNAs. Despite differences in IDH mutation status and significant intratumoral heterogeneity, the profiled tumor cells shared a common chromatin structure defined by open regions enriched for nuclear factor 1 transcription factors (NFIA and NFIB). Silencing of NFIA or NFIB suppressed in vitro and in vivo growths of patient-derived glioblastomas and G4 IDHm astrocytoma models. These findings suggest that despite distinct genotypes and cell states, glioblastoma/G4 astrocytoma cells share dependency on core transcriptional programs, yielding an attractive platform for addressing therapeutic challenges associated with intratumoral heterogeneity.


Asunto(s)
Astrocitoma , Neoplasias Encefálicas , Glioblastoma , Adulto , Humanos , Glioblastoma/genética , Glioblastoma/patología , Cromatina/genética , Transcriptoma , Astrocitoma/genética , Astrocitoma/patología , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patología , Mutación , Isocitrato Deshidrogenasa/genética , Isocitrato Deshidrogenasa/metabolismo
5.
Cell Res ; 32(11): 1008-1021, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-36207411

RESUMEN

Loss of heterochromatin has been implicated as a cause of pre-mature aging and age-associated decline in organ functions in mammals; however, the specific cell types and gene loci affected by this type of epigenetic change have remained unclear. To address this knowledge gap, we probed chromatin accessibility at single-cell resolution in the brains, hearts, skeletal muscles, and bone marrows from young, middle-aged, and old mice, and assessed age-associated changes at 353,126 candidate cis-regulatory elements (cCREs) across 32 major cell types. Unexpectedly, we detected increased chromatin accessibility within specific heterochromatin domains in old mouse excitatory neurons. The gain of chromatin accessibility at these genomic loci was accompanied by the cell-type-specific loss of heterochromatin and activation of LINE1 elements. Immunostaining further confirmed the loss of the heterochromatin mark H3K9me3 in the excitatory neurons but not in inhibitory neurons or glial cells. Our results reveal the cell-type-specific changes in chromatin landscapes in old mice and shed light on the scope of heterochromatin loss in mammalian aging.


Asunto(s)
Epigenoma , Heterocromatina , Ratones , Animales , Cromatina , Neuronas , Encéfalo , Mamíferos/genética
7.
Cell ; 184(24): 5985-6001.e19, 2021 11 24.
Artículo en Inglés | MEDLINE | ID: mdl-34774128

RESUMEN

Current catalogs of regulatory sequences in the human genome are still incomplete and lack cell type resolution. To profile the activity of gene regulatory elements in diverse cell types and tissues in the human body, we applied single-cell chromatin accessibility assays to 30 adult human tissue types from multiple donors. We integrated these datasets with previous single-cell chromatin accessibility data from 15 fetal tissue types to reveal the status of open chromatin for ∼1.2 million candidate cis-regulatory elements (cCREs) in 222 distinct cell types comprised of >1.3 million nuclei. We used these chromatin accessibility maps to delineate cell-type-specificity of fetal and adult human cCREs and to systematically interpret the noncoding variants associated with complex human traits and diseases. This rich resource provides a foundation for the analysis of gene regulatory programs in human cell types across tissues, life stages, and organ systems.


Asunto(s)
Cromatina/metabolismo , Genoma Humano , Análisis de la Célula Individual , Adulto , Análisis por Conglomerados , Feto/metabolismo , Variación Genética , Estudio de Asociación del Genoma Completo , Humanos , Especificidad de Órganos , Filogenia , Secuencias Reguladoras de Ácidos Nucleicos/genética , Factores de Riesgo
8.
Molecules ; 26(21)2021 Nov 02.
Artículo en Inglés | MEDLINE | ID: mdl-34771043

RESUMEN

Neural stem cells (NSCs) exist in the central nervous system of adult animals and capable of self-replication. NSCs have two basic functions, namely the proliferation ability and the potential for multi-directional differentiation. In this study, based on the bioassay-guided fractionation, we aim to screen active components in Cuscuta chinensis to promote the proliferation of NSCs. CCK-8 assays were used as an active detection method to track the active components. On the basis of isolating active fraction and monomer compounds, the structures of these were identified by LC-MS and (1H, 13C) NMR. Moreover, active components were verified by pharmacodynamics and network pharmacology. The system solvent extraction method combined with the traditional isolation method were used to ensure that the fraction TSZE-EA-G6 of Cuscuta chinensis exhibited the highest activity. Seven chemical components were identified from the TSZE-EA-G6 fraction by UPLC-QE-Orbitrap-MS technology, which were 4-O-p-coumarinic acid, chlorogenic acid, 5-O-p-coumarinic acid, hyperoside, astragalin, isochlorogenic acid C, and quercetin-3-O-galactose-7-O-glucoside. Using different chromatographic techniques, five compounds were isolated in TSZE-EA-G6 and identified as kaempferol, kaempferol-3-O-glucoside (astragalin), quercetin-3-O-galactoside (hyperoside), chlorogenic acid, and sucrose. The activity study of these five compounds showed that the proliferation rate of kaempferol had the highest effects; at a certain concentration (25 µg/mL, 3.12 µg/mL), the proliferation rate could reach 87.44% and 59.59%, respectively. Furthermore, research results using network pharmacology techniques verified that kaempferol had an activity of promoting NSCs proliferation and the activity of flavonoid aglycones might be greater than that of flavonoid glycosides. In conclusion, this research shows that kaempferol is the active component in Cuscuta chinensis to promote the proliferation of NSCs.


Asunto(s)
Cuscuta/química , Medicamentos Herbarios Chinos/farmacología , Células-Madre Neurales/efectos de los fármacos , Animales , Proliferación Celular/efectos de los fármacos , Fraccionamiento Químico , Cromatografía Líquida de Alta Presión , Relación Dosis-Respuesta a Droga , Evaluación Preclínica de Medicamentos , Medicamentos Herbarios Chinos/química , Medicamentos Herbarios Chinos/aislamiento & purificación , Femenino , Espectrometría de Masas , Células-Madre Neurales/citología , Ratas
9.
Nature ; 598(7879): 120-128, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34616061

RESUMEN

Mammalian brain cells show remarkable diversity in gene expression, anatomy and function, yet the regulatory DNA landscape underlying this extensive heterogeneity is poorly understood. Here we carry out a comprehensive assessment of the epigenomes of mouse brain cell types by applying single-nucleus DNA methylation sequencing1,2 to profile 103,982 nuclei (including 95,815 neurons and 8,167 non-neuronal cells) from 45 regions of the mouse cortex, hippocampus, striatum, pallidum and olfactory areas. We identified 161 cell clusters with distinct spatial locations and projection targets. We constructed taxonomies of these epigenetic types, annotated with signature genes, regulatory elements and transcription factors. These features indicate the potential regulatory landscape supporting the assignment of putative cell types and reveal repetitive usage of regulators in excitatory and inhibitory cells for determining subtypes. The DNA methylation landscape of excitatory neurons in the cortex and hippocampus varied continuously along spatial gradients. Using this deep dataset, we constructed an artificial neural network model that precisely predicts single neuron cell-type identity and brain area spatial location. Integration of high-resolution DNA methylomes with single-nucleus chromatin accessibility data3 enabled prediction of high-confidence enhancer-gene interactions for all identified cell types, which were subsequently validated by cell-type-specific chromatin conformation capture experiments4. By combining multi-omic datasets (DNA methylation, chromatin contacts, and open chromatin) from single nuclei and annotating the regulatory genome of hundreds of cell types in the mouse brain, our DNA methylation atlas establishes the epigenetic basis for neuronal diversity and spatial organization throughout the mouse cerebrum.


Asunto(s)
Encéfalo/citología , Metilación de ADN , Epigenoma , Epigenómica , Neuronas/clasificación , Neuronas/metabolismo , Análisis de la Célula Individual , Animales , Atlas como Asunto , Encéfalo/metabolismo , Cromatina/química , Cromatina/genética , Cromatina/metabolismo , Citosina/química , Citosina/metabolismo , Conjuntos de Datos como Asunto , Giro Dentado/citología , Elementos de Facilitación Genéticos/genética , Perfilación de la Expresión Génica , Hipocampo/citología , Hipocampo/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Modelos Biológicos , Vías Nerviosas , Neuronas/citología
10.
Nature ; 598(7879): 111-119, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34616062

RESUMEN

The primary motor cortex (M1) is essential for voluntary fine-motor control and is functionally conserved across mammals1. Here, using high-throughput transcriptomic and epigenomic profiling of more than 450,000 single nuclei in humans, marmoset monkeys and mice, we demonstrate a broadly conserved cellular makeup of this region, with similarities that mirror evolutionary distance and are consistent between the transcriptome and epigenome. The core conserved molecular identities of neuronal and non-neuronal cell types allow us to generate a cross-species consensus classification of cell types, and to infer conserved properties of cell types across species. Despite the overall conservation, however, many species-dependent specializations are apparent, including differences in cell-type proportions, gene expression, DNA methylation and chromatin state. Few cell-type marker genes are conserved across species, revealing a short list of candidate genes and regulatory mechanisms that are responsible for conserved features of homologous cell types, such as the GABAergic chandelier cells. This consensus transcriptomic classification allows us to use patch-seq (a combination of whole-cell patch-clamp recordings, RNA sequencing and morphological characterization) to identify corticospinal Betz cells from layer 5 in non-human primates and humans, and to characterize their highly specialized physiology and anatomy. These findings highlight the robust molecular underpinnings of cell-type diversity in M1 across mammals, and point to the genes and regulatory pathways responsible for the functional identity of cell types and their species-specific adaptations.


Asunto(s)
Corteza Motora/citología , Neuronas/clasificación , Análisis de la Célula Individual , Animales , Atlas como Asunto , Callithrix/genética , Epigénesis Genética , Epigenómica , Femenino , Neuronas GABAérgicas/citología , Neuronas GABAérgicas/metabolismo , Perfilación de la Expresión Génica , Glutamatos/metabolismo , Humanos , Hibridación Fluorescente in Situ , Masculino , Ratones , Persona de Mediana Edad , Corteza Motora/anatomía & histología , Neuronas/citología , Neuronas/metabolismo , Especificidad de Órganos , Filogenia , Especificidad de la Especie , Transcriptoma
11.
Nature ; 598(7879): 103-110, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34616066

RESUMEN

Single-cell transcriptomics can provide quantitative molecular signatures for large, unbiased samples of the diverse cell types in the brain1-3. With the proliferation of multi-omics datasets, a major challenge is to validate and integrate results into a biological understanding of cell-type organization. Here we generated transcriptomes and epigenomes from more than 500,000 individual cells in the mouse primary motor cortex, a structure that has an evolutionarily conserved role in locomotion. We developed computational and statistical methods to integrate multimodal data and quantitatively validate cell-type reproducibility. The resulting reference atlas-containing over 56 neuronal cell types that are highly replicable across analysis methods, sequencing technologies and modalities-is a comprehensive molecular and genomic account of the diverse neuronal and non-neuronal cell types in the mouse primary motor cortex. The atlas includes a population of excitatory neurons that resemble pyramidal cells in layer 4 in other cortical regions4. We further discovered thousands of concordant marker genes and gene regulatory elements for these cell types. Our results highlight the complex molecular regulation of cell types in the brain and will directly enable the design of reagents to target specific cell types in the mouse primary motor cortex for functional analysis.


Asunto(s)
Epigenómica , Perfilación de la Expresión Génica , Corteza Motora/citología , Neuronas/clasificación , Análisis de la Célula Individual , Transcriptoma , Animales , Atlas como Asunto , Conjuntos de Datos como Asunto , Epigénesis Genética , Femenino , Masculino , Ratones , Corteza Motora/anatomía & histología , Neuronas/citología , Neuronas/metabolismo , Especificidad de Órganos , Reproducibilidad de los Resultados
12.
Nature ; 598(7879): 129-136, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34616068

RESUMEN

The mammalian cerebrum performs high-level sensory perception, motor control and cognitive functions through highly specialized cortical and subcortical structures1. Recent surveys of mouse and human brains with single-cell transcriptomics2-6 and high-throughput imaging technologies7,8 have uncovered hundreds of neural cell types distributed in different brain regions, but the transcriptional regulatory programs that are responsible for the unique identity and function of each cell type remain unknown. Here we probe the accessible chromatin in more than 800,000 individual nuclei from 45 regions that span the adult mouse isocortex, olfactory bulb, hippocampus and cerebral nuclei, and use the resulting data to map the state of 491,818 candidate cis-regulatory DNA elements in 160 distinct cell types. We find high specificity of spatial distribution for not only excitatory neurons, but also most classes of inhibitory neurons and a subset of glial cell types. We characterize the gene regulatory sequences associated with the regional specificity within these cell types. We further link a considerable fraction of the cis-regulatory elements to putative target genes expressed in diverse cerebral cell types and predict transcriptional regulators that are involved in a broad spectrum of molecular and cellular pathways in different neuronal and glial cell populations. Our results provide a foundation for comprehensive analysis of gene regulatory programs of the mammalian brain and assist in the interpretation of noncoding risk variants associated with various neurological diseases and traits in humans.


Asunto(s)
Cerebro/citología , Cerebro/metabolismo , Secuencias Reguladoras de Ácidos Nucleicos/genética , Animales , Atlas como Asunto , Cromatina/química , Cromatina/genética , Cromatina/metabolismo , Ensamble y Desensamble de Cromatina , Regulación de la Expresión Génica , Predisposición Genética a la Enfermedad/genética , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Enfermedades del Sistema Nervioso/genética , Neuroglía/clasificación , Neuroglía/metabolismo , Neuronas/clasificación , Neuronas/metabolismo , Análisis de Secuencia de ADN , Análisis de la Célula Individual
13.
Ther Adv Chronic Dis ; 12: 20406223211028768, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34285789

RESUMEN

BACKGROUND: Chronic obstructive pulmonary disease (COPD) is characterized by persistent respiratory symptoms and dyspnea, as well as an increase in the number of leukocytes in the airways, lungs, and pulmonary vessels. A 'One size fits all' approach to COPD patients with different clinical features may be considered outdated. The following are the two major objectives of this meta-analysis: the first is to determine if blood eosinophil counts (BEC) can serve as a prognostic biomarker of COPD outcomes, and the second is to determine which level of BEC is effective for inhaled corticosteroid (ICS) treatment. METHODS: We searched articles published before 15 May 2021 in the following four electronic databases: Web of Science, Cochrane Library, EMBASE, and PubMed. RESULTS: A total of 42 studies, comprising a sampling of 188,710 subjects, were summarized and compared in this meta-analysis. The rate ratio (RR) of exacerbations of COPD (ECOPD) between ICS and non-ICS treatment was statistically significant for the COPD patients with a baseline BEC ⩾ 2% or ⩾ 200 cells/µl, RR = 0.82 (0.73, 0.93) or 0.79 (0.70, 0.89) respectively, while the RR of ECOPD between ICS and non-ICS treatment was statistically insignificant for the COPD patients with baseline BEC < 2% or <200 cells/µl, RR = 0.97 (0.87, 1.08) or 0.97 (0.86, 1.08), suggested that ICS therapy was beneficial to the improvement of ECOPD in patients with a baseline BEC ⩾ 2% or BEC ⩾ 200 cells/µl. CONCLUSION: Our research shows that a BEC ⩾ 200 cells/µl or ⩾2% is likely to become the cutoff value of ICS treatment for ECOPD. Moreover, we believe that the baseline BEC can be used as a biomarker for predicting ECOPD. The stability of BEC requires special attention.

14.
Sci Adv ; 7(20)2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33990324

RESUMEN

Misregulated gene expression in human hearts can result in cardiovascular diseases that are leading causes of mortality worldwide. However, the limited information on the genomic location of candidate cis-regulatory elements (cCREs) such as enhancers and promoters in distinct cardiac cell types has restricted the understanding of these diseases. Here, we defined >287,000 cCREs in the four chambers of the human heart at single-cell resolution, which revealed cCREs and candidate transcription factors associated with cardiac cell types in a region-dependent manner and during heart failure. We further found cardiovascular disease-associated genetic variants enriched within these cCREs including 38 candidate causal atrial fibrillation variants localized to cardiomyocyte cCREs. Additional functional studies revealed that two of these variants affect a cCRE controlling KCNH2/HERG expression and action potential repolarization. Overall, this atlas of human cardiac cCREs provides the foundation for illuminating cell type-specific gene regulation in human hearts during health and disease.


Asunto(s)
Corazón , Secuencias Reguladoras de Ácidos Nucleicos , Humanos , Regiones Promotoras Genéticas , Secuencias Reguladoras de Ácidos Nucleicos/genética , Factores de Transcripción/metabolismo
15.
Nat Commun ; 12(1): 1337, 2021 02 26.
Artículo en Inglés | MEDLINE | ID: mdl-33637727

RESUMEN

Identification of the cis-regulatory elements controlling cell-type specific gene expression patterns is essential for understanding the origin of cellular diversity. Conventional assays to map regulatory elements via open chromatin analysis of primary tissues is hindered by sample heterogeneity. Single cell analysis of accessible chromatin (scATAC-seq) can overcome this limitation. However, the high-level noise of each single cell profile and the large volume of data pose unique computational challenges. Here, we introduce SnapATAC, a software package for analyzing scATAC-seq datasets. SnapATAC dissects cellular heterogeneity in an unbiased manner and map the trajectories of cellular states. Using the Nyström method, SnapATAC can process data from up to a million cells. Furthermore, SnapATAC incorporates existing tools into a comprehensive package for analyzing single cell ATAC-seq dataset. As demonstration of its utility, SnapATAC is applied to 55,592 single-nucleus ATAC-seq profiles from the mouse secondary motor cortex. The analysis reveals ~370,000 candidate regulatory elements in 31 distinct cell populations in this brain region and inferred candidate cell-type specific transcriptional regulators.


Asunto(s)
Secuenciación de Inmunoprecipitación de Cromatina/métodos , Análisis de la Célula Individual/métodos , Animales , Cromatina , Biología Computacional , Epigenómica , Masculino , Ratones , Ratones Endogámicos C57BL , Corteza Motora , Análisis de Secuencia de ADN/métodos
16.
JCI Insight ; 6(2)2021 01 25.
Artículo en Inglés | MEDLINE | ID: mdl-33320836

RESUMEN

The G/T transversion rs35705950, located approximately 3 kb upstream of the MUC5B start site, is the cardinal risk factor for idiopathic pulmonary fibrosis (IPF). Here, we investigate the function and chromatin structure of this -3 kb region and provide evidence that it functions as a classically defined enhancer subject to epigenetic programming. We use nascent transcript analysis to show that RNA polymerase II loads within 10 bp of the G/T transversion site, definitively establishing enhancer function for the region. By integrating Assay for Transposase-Accessible Chromatin using sequencing (ATAC-seq) analysis of fresh and cultured human airway epithelial cells with nuclease sensitivity data, we demonstrate that this region is in accessible chromatin that affects the expression of MUC5B. Through applying paired single-nucleus RNA- and ATAC-seq to frozen tissue from IPF lungs, we extend these findings directly to disease, with results indicating that epigenetic programming of the -3 kb enhancer in IPF occurs in both MUC5B-expressing and nonexpressing lineages. In aggregate, our results indicate that the MUC5B-associated variant rs35705950 resides within an enhancer that is subject to epigenetic remodeling and contributes to pathologic misexpression in IPF.


Asunto(s)
Fibrosis Pulmonar Idiopática/genética , Mucina 5B/genética , Células A549 , Sitios de Unión/genética , Línea Celular , Cromatina/genética , Cromatina/metabolismo , Elementos de Facilitación Genéticos , Epigénesis Genética , Mutación con Ganancia de Función , Regulación de la Expresión Génica , Predisposición Genética a la Enfermedad , Humanos , Fibrosis Pulmonar Idiopática/metabolismo , Pulmón/metabolismo , Modelos Genéticos , Polimorfismo de Nucleótido Simple , Proteínas Proto-Oncogénicas c-ets/metabolismo , ARN Polimerasa II/metabolismo , Factor de Transcripción STAT3/metabolismo
17.
Elife ; 92020 11 09.
Artículo en Inglés | MEDLINE | ID: mdl-33164753

RESUMEN

Respiratory failure associated with COVID-19 has placed focus on the lungs. Here, we present single-nucleus accessible chromatin profiles of 90,980 nuclei and matched single-nucleus transcriptomes of 46,500 nuclei in non-diseased lungs from donors of ~30 weeks gestation,~3 years and ~30 years. We mapped candidate cis-regulatory elements (cCREs) and linked them to putative target genes. We identified distal cCREs with age-increased activity linked to SARS-CoV-2 host entry gene TMPRSS2 in alveolar type 2 cells, which had immune regulatory signatures and harbored variants associated with respiratory traits. At the 3p21.31 COVID-19 risk locus, a candidate variant overlapped a distal cCRE linked to SLC6A20, a gene expressed in alveolar cells and with known functional association with the SARS-CoV-2 receptor ACE2. Our findings provide insight into regulatory logic underlying genes implicated in COVID-19 in individual lung cell types across age. More broadly, these datasets will facilitate interpretation of risk loci for lung diseases.


Asunto(s)
COVID-19/genética , COVID-19/virología , Interacciones Microbiota-Huesped/genética , Pulmón/metabolismo , Pulmón/virología , Adulto , Factores de Edad , Células Epiteliales Alveolares/clasificación , Células Epiteliales Alveolares/metabolismo , Células Epiteliales Alveolares/virología , Enzima Convertidora de Angiotensina 2/genética , Enzima Convertidora de Angiotensina 2/metabolismo , COVID-19/metabolismo , Preescolar , Mapeo Cromosómico , Perfilación de la Expresión Génica , Variación Genética , Interacciones Microbiota-Huesped/fisiología , Humanos , Recién Nacido , Proteínas de Transporte de Membrana/genética , Proteínas de Transporte de Membrana/metabolismo , Pandemias , Receptores Virales/genética , Receptores Virales/metabolismo , SARS-CoV-2/genética , SARS-CoV-2/patogenicidad , Análisis de la Célula Individual , Internalización del Virus
18.
Cell Chem Biol ; 27(9): 1140-1150.e4, 2020 09 17.
Artículo en Inglés | MEDLINE | ID: mdl-32649905

RESUMEN

Wnt/ß-catenin signaling regulates critical, context-dependent transcription in numerous physiological events. Among the well-documented mechanisms affecting Wnt/ß-catenin activity, modification of N-glycans by L-fucose is the newest and the least understood. Using a combination of Chinese hamster ovary cell mutants with different fucosylation levels and cell-surface fucose editing (in situ fucosylation [ISF]), we report that α(1-3)-fucosylation of N-acetylglucosamine (GlcNAc) in the Galß(1-4)-GlcNAc sequences of complex N-glycans modulates Wnt/ß-catenin activity by regulating the endocytosis of low-density lipoprotein receptor-related protein 6 (LRP6). Pulse-chase experiments reveal that ISF elevates endocytosis of lipid-raft-localized LRP6, leading to the suppression of Wnt/ß-catenin signaling. Remarkably, Wnt activity decreased by ISF is fully reversed by the exogenously added fucose. The combined data show that in situ cell-surface fucosylation can be exploited to regulate a specific signaling pathway via endocytosis promoted by a fucose-binding protein, thereby linking glycosylation of a receptor with its intracellular signaling.


Asunto(s)
Endocitosis , Fucosa/metabolismo , Proteína-6 Relacionada a Receptor de Lipoproteína de Baja Densidad/metabolismo , Vía de Señalización Wnt , Animales , Células CHO , Línea Celular , Cricetinae , Cricetulus , Embrión no Mamífero/metabolismo , Endocitosis/efectos de los fármacos , Fucosa/farmacología , Glicosilación , Humanos , Proteína-6 Relacionada a Receptor de Lipoproteína de Baja Densidad/genética , Microdominios de Membrana/metabolismo , Proteínas de Transporte de Monosacáridos/genética , Proteínas de Transporte de Monosacáridos/metabolismo , Proteínas Wnt/antagonistas & inhibidores , Proteínas Wnt/metabolismo , Pez Cebra/crecimiento & desarrollo , Pez Cebra/metabolismo , beta Catenina/metabolismo
19.
Nat Cell Biol ; 22(4): 487-497, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32231307

RESUMEN

During mouse embryonic development, pluripotent cells rapidly divide and diversify, yet the regulatory programs that define the cell repertoire for each organ remain ill-defined. To delineate comprehensive chromatin landscapes during early organogenesis, we mapped chromatin accessibility in 19,453 single nuclei from mouse embryos at 8.25 days post-fertilization. Identification of cell-type-specific regions of open chromatin pinpointed two TAL1-bound endothelial enhancers, which we validated using transgenic mouse assays. Integrated gene expression and transcription factor motif enrichment analyses highlighted cell-type-specific transcriptional regulators. Subsequent in vivo experiments in zebrafish revealed a role for the ETS factor FEV in endothelial identity downstream of ETV2 (Etsrp in zebrafish). Concerted in vivo validation experiments in mouse and zebrafish thus illustrate how single-cell open chromatin maps, representative of a mammalian embryo, provide access to the regulatory blueprint for mammalian organogenesis.


Asunto(s)
Cromatina/química , Células Endoteliales/metabolismo , Elementos de Facilitación Genéticos , Regulación del Desarrollo de la Expresión Génica , Organogénesis/genética , Proteína 1 de la Leucemia Linfocítica T Aguda/genética , Animales , Linaje de la Célula/genética , Núcleo Celular/genética , Núcleo Celular/metabolismo , Cromatina/metabolismo , Embrión de Mamíferos , Embrión no Mamífero , Desarrollo Embrionario , Células Endoteliales/citología , Perfilación de la Expresión Génica , Ratones , Ratones Transgénicos , Especificidad de Órganos , Unión Proteica , Análisis de la Célula Individual , Proteína 1 de la Leucemia Linfocítica T Aguda/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Transcripción Genética , Pez Cebra , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo
20.
Chem Sci ; 11(2): 474-481, 2020 Jan 14.
Artículo en Inglés | MEDLINE | ID: mdl-32190267

RESUMEN

Printable mesoscopic perovskite solar cells are usually fabricated by drop-casting perovskite precursor solution on a screen-printed mesoporous TiO2/ZrO2/carbon triple-layer followed by thermal annealing. They have attracted much attention due to their simple fabrication process and remarkable stability. However, challenges lie in how to achieve complete pore fillings of perovskites in the meso-pores and to obtain high-quality perovskite crystals. Here, we report an in situ crystal transfer (ICT) process based on gas-solid interaction to deposit perovskite CH3NH3PbI3 absorber in the scaffold. CH3NH3PbI3 single crystals are first transformed into a liquid phase via exposure to methylamine gas flow. After complete infiltration into the nano-structured scaffolds, the liquid phase is converted back to the solid phase with reduction of methylamine gas partial pressure, maintaining the high-quality of CH3NH3PbI3 single crystals. Compared with the conventional drop-casting method, the ICT method effectively leads to interconnected morphology and prolongs the charge-carrier lifetime (from ∼37.52 ns to ∼110.85 ns) of the perovskite absorber in the scaffold. As a result, the devices can deliver a power conversion efficiency of 15.89%, which is attributed to the suppressed charge recombination and correspondingly enhanced open-circuit voltage of 0.98 V.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA