Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 93
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 180(4): 764-779.e20, 2020 02 20.
Artículo en Inglés | MEDLINE | ID: mdl-32059779

RESUMEN

The heterogeneity of endothelial cells (ECs) across tissues remains incompletely inventoried. We constructed an atlas of >32,000 single-EC transcriptomes from 11 mouse tissues and identified 78 EC subclusters, including Aqp7+ intestinal capillaries and angiogenic ECs in healthy tissues. ECs from brain/testis, liver/spleen, small intestine/colon, and skeletal muscle/heart pairwise expressed partially overlapping marker genes. Arterial, venous, and lymphatic ECs shared more markers in more tissues than did heterogeneous capillary ECs. ECs from different vascular beds (arteries, capillaries, veins, lymphatics) exhibited transcriptome similarity across tissues, but the tissue (rather than the vessel) type contributed to the EC heterogeneity. Metabolic transcriptome analysis revealed a similar tissue-grouping phenomenon of ECs and heterogeneous metabolic gene signatures in ECs between tissues and between vascular beds within a single tissue in a tissue-type-dependent pattern. The EC atlas taxonomy enabled identification of EC subclusters in public scRNA-seq datasets and provides a powerful discovery tool and resource value.


Asunto(s)
Células Endoteliales/metabolismo , Análisis de la Célula Individual , Transcriptoma , Animales , Encéfalo/citología , Sistema Cardiovascular/citología , Células Endoteliales/clasificación , Células Endoteliales/citología , Tracto Gastrointestinal/citología , Masculino , Ratones , Ratones Endogámicos C57BL , Músculos/citología , Especificidad de Órganos , RNA-Seq , Testículo/citología
2.
Cell ; 174(4): 982-998.e20, 2018 08 09.
Artículo en Inglés | MEDLINE | ID: mdl-29909982

RESUMEN

The diversity of cell types and regulatory states in the brain, and how these change during aging, remains largely unknown. We present a single-cell transcriptome atlas of the entire adult Drosophila melanogaster brain sampled across its lifespan. Cell clustering identified 87 initial cell clusters that are further subclustered and validated by targeted cell-sorting. Our data show high granularity and identify a wide range of cell types. Gene network analyses using SCENIC revealed regulatory heterogeneity linked to energy consumption. During aging, RNA content declines exponentially without affecting neuronal identity in old brains. This single-cell brain atlas covers nearly all cells in the normal brain and provides the tools to study cellular diversity alongside other Drosophila and mammalian single-cell datasets in our unique single-cell analysis platform: SCope (http://scope.aertslab.org). These results, together with SCope, allow comprehensive exploration of all transcriptional states of an entire aging brain.


Asunto(s)
Envejecimiento , Encéfalo/metabolismo , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Redes Reguladoras de Genes , Análisis de la Célula Individual/métodos , Transcriptoma , Animales , Drosophila melanogaster/fisiología , Femenino , Perfilación de la Expresión Génica , Masculino
3.
Nat Rev Genet ; 24(8): 494-515, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-36864178

RESUMEN

The joint analysis of the genome, epigenome, transcriptome, proteome and/or metabolome from single cells is transforming our understanding of cell biology in health and disease. In less than a decade, the field has seen tremendous technological revolutions that enable crucial new insights into the interplay between intracellular and intercellular molecular mechanisms that govern development, physiology and pathogenesis. In this Review, we highlight advances in the fast-developing field of single-cell and spatial multi-omics technologies (also known as multimodal omics approaches), and the computational strategies needed to integrate information across these molecular layers. We demonstrate their impact on fundamental cell biology and translational research, discuss current challenges and provide an outlook to the future.


Asunto(s)
Genoma , Multiómica , Transcriptoma , Metaboloma , Proteoma/genética , Análisis de la Célula Individual
4.
Nature ; 579(7797): 111-117, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-32103177

RESUMEN

The avascular nature of cartilage makes it a unique tissue1-4, but whether and how the absence of nutrient supply regulates chondrogenesis remain unknown. Here we show that obstruction of vascular invasion during bone healing favours chondrogenic over osteogenic differentiation of skeletal progenitor cells. Unexpectedly, this process is driven by a decreased availability of extracellular lipids. When lipids are scarce, skeletal progenitors activate forkhead box O (FOXO) transcription factors, which bind to the Sox9 promoter and increase its expression. Besides initiating chondrogenesis, SOX9 acts as a regulator of cellular metabolism by suppressing oxidation of fatty acids, and thus adapts the cells to an avascular life. Our results define lipid scarcity as an important determinant of chondrogenic commitment, reveal a role for FOXO transcription factors during lipid starvation, and identify SOX9 as a critical metabolic mediator. These data highlight the importance of the nutritional microenvironment in the specification of skeletal cell fate.


Asunto(s)
Huesos/citología , Microambiente Celular , Condrogénesis , Metabolismo de los Lípidos , Factor de Transcripción SOX9/metabolismo , Células Madre/citología , Células Madre/metabolismo , Animales , Huesos/irrigación sanguínea , Condrocitos/citología , Condrocitos/metabolismo , Ácidos Grasos/metabolismo , Femenino , Privación de Alimentos , Factores de Transcripción Forkhead/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Osteogénesis , Oxidación-Reducción , Factor de Transcripción SOX9/genética , Transducción de Señal , Cicatrización de Heridas
5.
Nature ; 583(7817): 615-619, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32494007

RESUMEN

Pneumonia resulting from infection is one of the leading causes of death worldwide. Pulmonary infection by the respiratory syncytial virus (RSV) is a large burden on human health, for which there are few therapeutic options1. RSV targets ciliated epithelial cells in the airways, but how viruses such as RSV interact with receptors on these cells is not understood. Nucleolin is an entry coreceptor for RSV2 and also mediates the cellular entry of influenza, the parainfluenza virus, some enteroviruses and the bacterium that causes tularaemia3,4. Here we show a mechanism of RSV entry into cells in which outside-in signalling, involving binding of the prefusion RSV-F glycoprotein with the insulin-like growth factor-1 receptor, triggers the activation of protein kinase C zeta (PKCζ). This cellular signalling cascade recruits nucleolin from the nuclei of cells to the plasma membrane, where it also binds to RSV-F on virions. We find that inhibiting PKCζ activation prevents the trafficking of nucleolin to RSV particles on airway organoid cultures, and reduces viral replication and pathology in RSV-infected mice. These findings reveal a mechanism of virus entry in which receptor engagement and signal transduction bring the coreceptor to viral particles at the cell surface, and could form the basis of new therapeutics to treat RSV infection.


Asunto(s)
Receptor IGF Tipo 1/metabolismo , Receptores Virales/metabolismo , Virus Sincitiales Respiratorios/metabolismo , Internalización del Virus , Línea Celular , Núcleo Celular/metabolismo , Activación Enzimática , Humanos , Fusión de Membrana/efectos de los fármacos , Fosfoproteínas/metabolismo , Unión Proteica , Proteína Quinasa C/antagonistas & inhibidores , Proteína Quinasa C/metabolismo , Proteínas de Unión al ARN/metabolismo , Receptor IGF Tipo 1/antagonistas & inhibidores , Virus Sincitiales Respiratorios/efectos de los fármacos , Virus Sincitiales Respiratorios/patogenicidad , Virus Sincitiales Respiratorios/fisiología , Carga Viral/efectos de los fármacos , Internalización del Virus/efectos de los fármacos , Nucleolina
7.
Semin Cancer Biol ; 98: 1-10, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38029868

RESUMEN

Hypoxia is intrinsic to tumours and contributes to malignancy and metastasis while hindering the efficiency of existing treatments. Epigenetic mechanisms play a crucial role in the regulation of hypoxic cancer cell programs, both in the initial phases of sensing the decrease in oxygen levels and during adaptation to chronic lack of oxygen. During the latter, the epigenetic regulation of tumour biology intersects with hypoxia-sensitive transcription factors in a complex network of gene regulation that also involves metabolic reprogramming. Here, we review the current literature on the epigenetic control of gene programs in hypoxic cancer cells. We highlight common themes and features of such epigenetic remodelling and discuss their relevance for the development of therapeutic strategies.


Asunto(s)
Epigénesis Genética , Neoplasias , Humanos , Hipoxia de la Célula/genética , Hipoxia/genética , Hipoxia/metabolismo , Neoplasias/patología , Oxígeno/metabolismo
8.
Prenat Diagn ; 44(4): 418-421, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38047711

RESUMEN

Presymptomatic prediction of preeclampsia (PE) is crucial to enable early prophylactic treatment. Current screening tools are either complex or lack predictive value. We recently demonstrated that cell-free DNA methylation can be leveraged to predict early-onset PE in 57% at a 10% false positive rate. Importantly, this minimally invasive screening test can be implemented as an add-on to current widespread noninvasive prenatal aneuploidy screening. Here, we highlight the pitfalls and promising prospects of this research.


Asunto(s)
Ácidos Nucleicos Libres de Células , Preeclampsia , Embarazo , Femenino , Humanos , Preeclampsia/diagnóstico , Preeclampsia/genética , Preeclampsia/prevención & control , Salud Materna , Metilación de ADN , Aneuploidia
9.
PLoS Genet ; 17(7): e1009679, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34324492

RESUMEN

Numerous genetic studies have established a role for rare genomic variants in Congenital Heart Disease (CHD) at the copy number variation (CNV) and de novo variant (DNV) level. To identify novel haploinsufficient CHD disease genes, we performed an integrative analysis of CNVs and DNVs identified in probands with CHD including cases with sporadic thoracic aortic aneurysm. We assembled CNV data from 7,958 cases and 14,082 controls and performed a gene-wise analysis of the burden of rare genomic deletions in cases versus controls. In addition, we performed variation rate testing for DNVs identified in 2,489 parent-offspring trios. Our analysis revealed 21 genes which were significantly affected by rare CNVs and/or DNVs in probands. Fourteen of these genes have previously been associated with CHD while the remaining genes (FEZ1, MYO16, ARID1B, NALCN, WAC, KDM5B and WHSC1) have only been associated in small cases series or show new associations with CHD. In addition, a systems level analysis revealed affected protein-protein interaction networks involved in Notch signaling pathway, heart morphogenesis, DNA repair and cilia/centrosome function. Taken together, this approach highlights the importance of re-analyzing existing datasets to strengthen disease association and identify novel disease genes and pathways.


Asunto(s)
Variaciones en el Número de Copia de ADN/genética , Haploinsuficiencia/genética , Cardiopatías Congénitas/genética , Bases de Datos Genéticas , Expresión Génica/genética , Perfilación de la Expresión Génica/métodos , Predisposición Genética a la Enfermedad/genética , Genómica/métodos , Humanos , Canales Iónicos/genética , Proteínas de la Membrana/genética , Polimorfismo de Nucleótido Simple/genética , Transcriptoma/genética
11.
Nature ; 542(7639): 49-54, 2017 02 02.
Artículo en Inglés | MEDLINE | ID: mdl-28024299

RESUMEN

Lymphatic vessels are lined by lymphatic endothelial cells (LECs), and are critical for health. However, the role of metabolism in lymphatic development has not yet been elucidated. Here we report that in transgenic mouse models, LEC-specific loss of CPT1A, a rate-controlling enzyme in fatty acid ß-oxidation, impairs lymphatic development. LECs use fatty acid ß-oxidation to proliferate and for epigenetic regulation of lymphatic marker expression during LEC differentiation. Mechanistically, the transcription factor PROX1 upregulates CPT1A expression, which increases acetyl coenzyme A production dependent on fatty acid ß-oxidation. Acetyl coenzyme A is used by the histone acetyltransferase p300 to acetylate histones at lymphangiogenic genes. PROX1-p300 interaction facilitates preferential histone acetylation at PROX1-target genes. Through this metabolism-dependent mechanism, PROX1 mediates epigenetic changes that promote lymphangiogenesis. Notably, blockade of CPT1 enzymes inhibits injury-induced lymphangiogenesis, and replenishing acetyl coenzyme A by supplementing acetate rescues this process in vivo.


Asunto(s)
Ácidos Grasos/química , Ácidos Grasos/metabolismo , Linfangiogénesis , Vasos Linfáticos/citología , Vasos Linfáticos/metabolismo , Acetatos/farmacología , Acetilcoenzima A/metabolismo , Acetilación/efectos de los fármacos , Animales , Carnitina O-Palmitoiltransferasa/antagonistas & inhibidores , Carnitina O-Palmitoiltransferasa/genética , Carnitina O-Palmitoiltransferasa/metabolismo , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/genética , Células Endoteliales/citología , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Epigénesis Genética , Femenino , Histonas/metabolismo , Proteínas de Homeodominio/metabolismo , Células Endoteliales de la Vena Umbilical Humana , Humanos , Linfangiogénesis/efectos de los fármacos , Linfangiogénesis/genética , Vasos Linfáticos/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Oxidación-Reducción/efectos de los fármacos , Biosíntesis de Proteínas , Transcripción Genética , Proteínas Supresoras de Tumor/metabolismo , Arterias Umbilicales/citología , Regulación hacia Arriba
12.
Nature ; 537(7618): 63-68, 2016 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-27533040

RESUMEN

Hypermethylation of the promoters of tumour suppressor genes represses transcription of these genes, conferring growth advantages to cancer cells. How these changes arise is poorly understood. Here we show that the activity of oxygen-dependent ten-eleven translocation (TET) enzymes is reduced by tumour hypoxia in human and mouse cells. TET enzymes catalyse DNA demethylation through 5-methylcytosine oxidation. This reduction in activity occurs independently of hypoxia-associated alterations in TET expression, proliferation, metabolism, hypoxia-inducible factor activity or reactive oxygen species, and depends directly on oxygen shortage. Hypoxia-induced loss of TET activity increases hypermethylation at gene promoters in vitro. In patients, tumour suppressor gene promoters are markedly more methylated in hypoxic tumour tissue, independent of proliferation, stromal cell infiltration and tumour characteristics. Our data suggest that up to half of hypermethylation events are due to hypoxia, with these events conferring a selective advantage. Accordingly, increased hypoxia in mouse breast tumours increases hypermethylation, while restoration of tumour oxygenation abrogates this effect. Tumour hypoxia therefore acts as a novel regulator of DNA methylation.


Asunto(s)
Metilación de ADN , Proteínas de Unión al ADN/deficiencia , Oxigenasas de Función Mixta/deficiencia , Oxígeno/metabolismo , Proteínas Proto-Oncogénicas/deficiencia , Hipoxia Tumoral/fisiología , 5-Metilcitosina/metabolismo , Animales , Proliferación Celular , Metilación de ADN/efectos de los fármacos , Metilación de ADN/genética , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Dioxigenasas , Femenino , Silenciador del Gen/efectos de los fármacos , Genes Supresores de Tumor , Humanos , Masculino , Neoplasias Mamarias Animales/genética , Neoplasias Mamarias Animales/metabolismo , Neoplasias Mamarias Animales/patología , Ratones , Oxigenasas de Función Mixta/genética , Oxigenasas de Función Mixta/metabolismo , Oxidación-Reducción/efectos de los fármacos , Oxígeno/farmacología , Regiones Promotoras Genéticas/genética , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Células del Estroma/patología , Hipoxia Tumoral/efectos de los fármacos , Hipoxia Tumoral/genética
13.
Curr Opin Oncol ; 33(5): 476-484, 2021 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-34038918

RESUMEN

PURPOSE OF REVIEW: Although chemotherapeutics are considered as genotoxins for decades, their exact mutagenic impact on the genome of cancerous and normal cells of cancer patients was unknown for a long time. However, this knowledge is necessary to understand the long-term side effects of chemotherapy. A particular condition represents pregnant cancer patients being treated with chemotherapy. Since certain chemotherapeutics can cross the placenta, concerns exist about possible mutational effects on the fetus' genome with potential long-term health consequences. RECENT FINDINGS: Recent advances of next-generation sequencing (NGS) techniques have opened possibilities to explore the exact mutational footprint of chemotherapies in healthy tissue from treated cancer patients. However, the ultra-low frequency of chemotherapy-induced mutations, introduction of technical artefacts, and inaccessibility of normal tissue has posed important limitations. This review discusses five state-of-the-art approaches that were recently designed to overcome these drawbacks. SUMMARY: Results of the latest investigations give valuable insights into the genome-wide genotoxicity profile of frequently applied chemotherapies, with most of these drugs being associated with a signature of random base substitutions and small indels. Though these findings still might be limited to extrapolate to healthy tissue, they pave the way for studies on the origin of long-term chemotherapy-related adverse health effects.


Asunto(s)
Secuenciación de Nucleótidos de Alto Rendimiento , Neoplasias , Humanos , Mutación , Neoplasias/tratamiento farmacológico , Neoplasias/genética
14.
Mol Cancer ; 19(1): 141, 2020 09 11.
Artículo en Inglés | MEDLINE | ID: mdl-32917214

RESUMEN

Immunotherapy (IO) has revolutionized the therapy landscape of non-small cell lung cancer (NSCLC), significantly prolonging the overall survival (OS) of advanced stage patients. Over the recent years IO therapy has been broadly integrated into the first-line setting of non-oncogene driven NSCLC, either in combination with chemotherapy, or in selected patients with PD-L1high expression as monotherapy. Still, a significant proportion of patients suffer from disease progression. A better understanding of resistance mechanisms depicts a central goal to avoid or overcome IO resistance and to improve patient outcome.We here review major cellular and molecular pathways within the tumor microenvironment (TME) that may impact the evolution of IO resistance. We summarize upcoming treatment options after IO resistance including novel IO targets (e.g. RIG-I, STING) as well as interesting combinational approaches such as IO combined with anti-angiogenic agents or metabolic targets (e.g. IDO-1, adenosine signaling, arginase). By discussing the fundamental mode of action of IO within the TME, we aim to understand and manage IO resistance and to seed new ideas for effective therapeutic IO concepts.


Asunto(s)
Antígeno B7-H1/genética , Carcinoma de Pulmón de Células no Pequeñas/terapia , Proteína 58 DEAD Box/genética , Inmunoterapia/efectos adversos , Proteínas de la Membrana/genética , Receptores Inmunológicos/genética , Arginasa/genética , Antígeno B7-H1/antagonistas & inhibidores , Antígeno B7-H1/inmunología , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/inmunología , Proteína 58 DEAD Box/antagonistas & inhibidores , Proteína 58 DEAD Box/inmunología , Resistencia a Antineoplásicos/genética , Resistencia a Antineoplásicos/inmunología , Humanos , Indolamina-Pirrol 2,3,-Dioxigenasa/genética , Proteínas de la Membrana/antagonistas & inhibidores , Proteínas de la Membrana/inmunología , Receptores Inmunológicos/antagonistas & inhibidores , Receptores Inmunológicos/inmunología , Microambiente Tumoral/efectos de los fármacos , Microambiente Tumoral/inmunología
15.
Mol Cell ; 48(6): 849-62, 2012 Dec 28.
Artículo en Inglés | MEDLINE | ID: mdl-23219530

RESUMEN

Genome-wide DNA methylation reprogramming occurs in mouse primordial germ cells (PGCs) and preimplantation embryos, but the precise dynamics and biological outcomes are largely unknown. We have carried out whole-genome bisulfite sequencing (BS-Seq) and RNA-Seq across key stages from E6.5 epiblast to E16.5 PGCs. Global loss of methylation takes place during PGC expansion and migration with evidence for passive demethylation, but sequences that carry long-term epigenetic memory (imprints, CpG islands on the X chromosome, germline-specific genes) only become demethylated upon entry of PGCs into the gonads. The transcriptional profile of PGCs is tightly controlled despite global hypomethylation, with transient expression of the pluripotency network, suggesting that reprogramming and pluripotency are inextricably linked. Our results provide a framework for the understanding of the epigenetic ground state of pluripotency in the germline.


Asunto(s)
Metilación de ADN/genética , Genoma , Impresión Genómica , Células Germinativas/metabolismo , Transcriptoma , Animales , Islas de CpG , Femenino , Estratos Germinativos/citología , Masculino , Ratones , Ratones Endogámicos C57BL , Análisis de Secuencia de ADN , Transcripción Genética , Cromosoma X/genética , Cromosoma X/metabolismo
16.
Kidney Int ; 96(5): 1195-1204, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31530476

RESUMEN

During ageing, kidney function decreases due to renal tubular atrophy, interstitial fibrosis, glomerulosclerosis and arteriosclerosis. Recently, changes in DNA methylation were shown to contribute to various ageing processes. However, it is unknown whether such changes also contribute to age-related kidney dysfunction. To assess this, we profiled genome-wide changes in DNA methylation (over 800 000 CpG sites) in 95 renal biopsies obtained prior to kidney transplantation from donors aged 16 to 73 years. Donor age significantly associated with the methylation of 92 778 CpGs (false discovery rate under 0.05), corresponding to 10 285 differentially methylated regions. These regions were most frequently located in genes involved in the Wnt/beta-catenin signaling pathway. Using an independent cohort of 67 biopsies, we autonomously validated these findings. Interestingly, the methylation status of these 92 778 age-related CpGs was associated with glomerulosclerosis (34.4% of CpGs at a false discovery rate under 0.05) and interstitial fibrosis (0.9%) and graft function at one year after transplantation, but not with tubular atrophy and arteriosclerosis. No association was observed with any of these pathologies at the time of transplantation (0% at a false discovery rate under 0.05). Thus, age-associated changes in DNA methylation at the time of transplantation predict future injury of transplanted kidneys. Specifically, our epigenome-wide association study demonstrates that epigenetic renal ageing is implicated in progressive fibrosis in both the glomerulus and the interstitium.


Asunto(s)
Envejecimiento/metabolismo , Metilación de ADN , Riñón/metabolismo , Adolescente , Adulto , Anciano , Femenino , Fibrosis , Humanos , Riñón/patología , Trasplante de Riñón , Masculino , Persona de Mediana Edad , Nefroesclerosis/etiología , Vía de Señalización Wnt/genética , Adulto Joven
17.
Thorax ; 74(2): 132-140, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30366970

RESUMEN

BACKGROUND: Idiopathic pulmonary fibrosis (IPF) is a severe lung disease characterised by extensive pathological changes. The objective for this study was to identify the gene network and regulators underlying disease pathology in IPF and its association with lung function. METHODS: Lung Tissue Research Consortium dataset with 262 IPF and control subjects (GSE47460) was randomly divided into two non-overlapping groups for cross-validated differential gene expression analysis. Consensus weighted gene coexpression network analysis identified overlapping coexpressed gene modules between both IPF groups. Modules were correlated with lung function (diffusion capacity, DLCO; forced expiratory volume in 1 s, FEV1; forced vital capacity, FVC) and enrichment analyses used to identify biological function and transcription factors. Module correlation with miRNA data (GSE72967) identified associated regulators. Clinical relevance in IPF was assessed in a peripheral blood gene expression dataset (GSE93606) to identify modules related to survival. RESULTS: Correlation network analysis identified 16 modules in IPF. Upregulated modules were associated with cilia, DNA replication and repair, contractile fibres, B-cell and unfolded protein response, and extracellular matrix. Downregulated modules were associated with blood vessels, T-cell and interferon responses, leucocyte activation and degranulation, surfactant metabolism, and cellular metabolic and catabolic processes. Lung function correlated with nine modules (eight with DLCO, five with FVC). Intermodular network of transcription factors and miRNA showed clustering of fibrosis, immune response and contractile modules. The cilia-associated module was able to predict survival (p=0.0097) in an independent peripheral blood IPF cohort. CONCLUSIONS: We identified a correlation gene expression network with associated regulators in IPF that provides novel insight into the pathological process of this disease.


Asunto(s)
Redes Reguladoras de Genes/genética , Fibrosis Pulmonar Idiopática/genética , Pulmón/fisiopatología , Anciano , Femenino , Predisposición Genética a la Enfermedad , Humanos , Fibrosis Pulmonar Idiopática/mortalidad , Fibrosis Pulmonar Idiopática/fisiopatología , Masculino , Persona de Mediana Edad , Reproducibilidad de los Resultados , Pruebas de Función Respiratoria/métodos , Análisis de Supervivencia , Transcriptoma/genética
18.
J Am Soc Nephrol ; 29(5): 1566-1576, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29610404

RESUMEN

Background Ischemia during kidney transplant causes chronic allograft injury and adversely affects outcome, but the underlying mechanisms are incompletely understood. In tumors, oxygen shortage reduces the DNA demethylating activity of the ten-11 translocation (TET) enzymes, yielding hypermethylated genomes that promote tumor progression. We investigated whether ischemia similarly induces DNA hypermethylation in kidney transplants and contributes to chronic injury.Methods We profiled genome-wide DNA methylation in three cohorts of brain-dead donor kidney allograft biopsy specimens: a longitudinal cohort with paired biopsy specimens obtained at allograft procurement (preischemia; n=13), after implantation and reperfusion (postischemia; n=13), and at 3 or 12 months after transplant (n=5 each); a cross-sectional cohort with preimplantation biopsy specimens (n=82); and a cross-sectional cohort with postreperfusion biopsy specimens (n=46).Results Analysis of the paired preischemia and postischemia specimens revealed that methylation increased drastically in all allografts on ischemia. Hypermethylation was caused by loss of 5-hydroxymethylcytosine, the product of TET activity, and it was stable 1 year after transplant. In the preimplantation cohort, CpG hypermethylation directly correlated with ischemia time and for some CpGs, increased 2.6% per additional hour of ischemia. Hypermethylation preferentially affected and reduced the expression of genes involved in suppressing kidney injury and fibrosis. Moreover, CpG hypermethylation in preimplantation specimens predicted chronic injury, particularly fibrosis and glomerulosclerosis, 1 year after transplant. This finding was validated in the independent postreperfusion cohort, in which hypermethylation also predicted reduced allograft function 1 year after transplant, outperforming established clinical variables.Conclusions We highlight a novel epigenetic basis for ischemia-induced chronic allograft injury with biomarker potential.


Asunto(s)
Aloinjertos/patología , Aloinjertos/fisiopatología , Isquemia Fría/efectos adversos , Metilación de ADN , Isquemia/genética , Isquemia/metabolismo , Trasplante de Riñón , 5-Metilcitosina/análogos & derivados , 5-Metilcitosina/metabolismo , Adulto , Anciano , Aloinjertos/enzimología , Islas de CpG , Estudios Transversales , Proteínas de Unión al ADN/metabolismo , Dioxigenasas/metabolismo , Femenino , Fibrosis , Expresión Génica , Glomeruloesclerosis Focal y Segmentaria/etiología , Glomeruloesclerosis Focal y Segmentaria/genética , Humanos , Isquemia/complicaciones , Estudios Longitudinales , Masculino , Persona de Mediana Edad , Oxigenasas de Función Mixta/metabolismo , Periodo Posoperatorio , Valor Predictivo de las Pruebas , Periodo Preoperatorio , Proteínas Proto-Oncogénicas/metabolismo
19.
Am J Hum Genet ; 94(4): 574-85, 2014 04 03.
Artículo en Inglés | MEDLINE | ID: mdl-24702954

RESUMEN

Congenital heart defects (CHDs) are the most common birth defect worldwide and are a leading cause of neonatal mortality. Nonsyndromic atrioventricular septal defects (AVSDs) are an important subtype of CHDs for which the genetic architecture is poorly understood. We performed exome sequencing in 13 parent-offspring trios and 112 unrelated individuals with nonsyndromic AVSDs and identified five rare missense variants (two of which arose de novo) in the highly conserved gene NR2F2, a very significant enrichment (p = 7.7 × 10(-7)) compared to 5,194 control subjects. We identified three additional CHD-affected families with other variants in NR2F2 including a de novo balanced chromosomal translocation, a de novo substitution disrupting a splice donor site, and a 3 bp duplication that cosegregated in a multiplex family. NR2F2 encodes a pleiotropic developmental transcription factor, and decreased dosage of NR2F2 in mice has been shown to result in abnormal development of atrioventricular septa. Via luciferase assays, we showed that all six coding sequence variants observed in individuals significantly alter the activity of NR2F2 on target promoters.


Asunto(s)
Factor de Transcripción COUP II/genética , Cardiopatías Congénitas/genética , Animales , Sitios de Unión , Factor de Transcripción COUP II/metabolismo , Línea Celular , Exoma , Femenino , Humanos , Masculino , Ratones , Mutación Missense , Linaje , Estudios Prospectivos , Transcripción Genética
20.
N Engl J Med ; 381(9): e16, 2019 08 29.
Artículo en Inglés | MEDLINE | ID: mdl-31461603

Asunto(s)
Genómica , Riñón , Aloinjertos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA