Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 186(6): 1179-1194.e15, 2023 03 16.
Artículo en Inglés | MEDLINE | ID: mdl-36931245

RESUMEN

The human brain undergoes rapid development at mid-gestation from a pool of neural stem and progenitor cells (NSPCs) that give rise to the neurons, oligodendrocytes, and astrocytes of the mature brain. Functional study of these cell types has been hampered by a lack of precise purification methods. We describe a method for prospectively isolating ten distinct NSPC types from the developing human brain using cell-surface markers. CD24-THY1-/lo cells were enriched for radial glia, which robustly engrafted and differentiated into all three neural lineages in the mouse brain. THY1hi cells marked unipotent oligodendrocyte precursors committed to an oligodendroglial fate, and CD24+THY1-/lo cells marked committed excitatory and inhibitory neuronal lineages. Notably, we identify and functionally characterize a transcriptomically distinct THY1hiEGFRhiPDGFRA- bipotent glial progenitor cell (GPC), which is lineage-restricted to astrocytes and oligodendrocytes, but not to neurons. Our study provides a framework for the functional study of distinct cell types in human neurodevelopment.


Asunto(s)
Células-Madre Neurales , Ratones , Animales , Humanos , Células-Madre Neurales/metabolismo , Neuronas , Diferenciación Celular/fisiología , Neuroglía/metabolismo , Encéfalo , Astrocitos
2.
Nature ; 619(7971): 860-867, 2023 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-37468622

RESUMEN

Many cancers originate from stem or progenitor cells hijacked by somatic mutations that drive replication, exemplified by adenomatous transformation of pulmonary alveolar epithelial type II (AT2) cells1. Here we demonstrate a different scenario: expression of KRAS(G12D) in differentiated AT1 cells reprograms them slowly and asynchronously back into AT2 stem cells that go on to generate indolent tumours. Like human lepidic adenocarcinoma, the tumour cells slowly spread along alveolar walls in a non-destructive manner and have low ERK activity. We find that AT1 and AT2 cells act as distinct cells of origin and manifest divergent responses to concomitant WNT activation and KRAS(G12D) induction, which accelerates AT2-derived but inhibits AT1-derived adenoma proliferation. Augmentation of ERK activity in KRAS(G12D)-induced AT1 cells increases transformation efficiency, proliferation and progression from lepidic to mixed tumour histology. Overall, we have identified a new cell of origin for lung adenocarcinoma, the AT1 cell, which recapitulates features of human lepidic cancer. In so doing, we also uncover a capacity for oncogenic KRAS to reprogram a differentiated and quiescent cell back into its parent stem cell en route to adenomatous transformation. Our work further reveals that irrespective of a given cancer's current molecular profile and driver oncogene, the cell of origin exerts a pervasive and perduring influence on its subsequent behaviour.


Asunto(s)
Adenocarcinoma del Pulmón , Reprogramación Celular , Neoplasias Pulmonares , Proteínas Proto-Oncogénicas p21(ras) , Células Madre , Humanos , Adenocarcinoma del Pulmón/genética , Adenocarcinoma del Pulmón/patología , Reprogramación Celular/genética , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Proteínas Proto-Oncogénicas p21(ras)/genética , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Células Madre/metabolismo , Células Madre/patología , Quinasas MAP Reguladas por Señal Extracelular/metabolismo
3.
Cell ; 155(5): 1178-87, 2013 Nov 21.
Artículo en Inglés | MEDLINE | ID: mdl-24267896

RESUMEN

There are few substantive methods to measure the health of the immune system, and the connection between immune strength and the viral component of the microbiome is poorly understood. Organ transplant recipients are treated with posttransplant therapies that combine immunosuppressive and antiviral drugs, offering a window into the effects of immune modulation on the virome. We used sequencing of cell-free DNA in plasma to investigate drug-virome interactions in a cohort of organ transplant recipients (656 samples, 96 patients) and find that antivirals and immunosuppressants strongly affect the structure of the virome in plasma. We observe marked virome compositional dynamics at the onset of the therapy and find that the total viral load increases with immunosuppression, whereas the bacterial component of the microbiome remains largely unaffected. The data provide insight into the relationship between the human virome, the state of the immune system, and the effects of pharmacological treatment and offer a potential application of the virome state to predict immunocompetence.


Asunto(s)
Antivirales/uso terapéutico , Sangre/virología , Trasplante de Corazón , Inmunosupresores/uso terapéutico , Trasplante de Pulmón , Virus/aislamiento & purificación , Adulto , Profilaxis Antibiótica , Sangre/microbiología , Niño , ADN/sangre , ADN/genética , Humanos , Virus/clasificación
4.
Cell ; 155(3): 621-35, 2013 Oct 24.
Artículo en Inglés | MEDLINE | ID: mdl-24243019

RESUMEN

Direct lineage reprogramming is a promising approach for human disease modeling and regenerative medicine, with poorly understood mechanisms. Here, we reveal a hierarchical mechanism in the direct conversion of fibroblasts into induced neuronal (iN) cells mediated by the transcription factors Ascl1, Brn2, and Myt1l. Ascl1 acts as an "on-target" pioneer factor by immediately occupying most cognate genomic sites in fibroblasts. In contrast, Brn2 and Myt1l do not access fibroblast chromatin productively on their own; instead, Ascl1 recruits Brn2 to Ascl1 sites genome wide. A unique trivalent chromatin signature in the host cells predicts the permissiveness for Ascl1 pioneering activity among different cell types. Finally, we identified Zfp238 as a key Ascl1 target gene that can partially substitute for Ascl1 during iN cell reprogramming. Thus, a precise match between pioneer factors and the chromatin context at key target genes is determinative for transdifferentiation to neurons and likely other cell types.


Asunto(s)
Reprogramación Celular , Embrión de Mamíferos/citología , Fibroblastos/citología , Redes Reguladoras de Genes , Neuronas/citología , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Diferenciación Celular , Cromatina/metabolismo , Fibroblastos/metabolismo , Estudio de Asociación del Genoma Completo , Humanos , Ratones , Proteínas del Tejido Nervioso/metabolismo , Neuronas/metabolismo , Factores del Dominio POU/metabolismo , Proteínas Represoras/metabolismo , Factores de Transcripción/metabolismo
5.
Nature ; 603(7900): 309-314, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35236985

RESUMEN

The ability to slow or reverse biological ageing would have major implications for mitigating disease risk and maintaining vitality1. Although an increasing number of interventions show promise for rejuvenation2, their effectiveness on disparate cell types across the body and the molecular pathways susceptible to rejuvenation remain largely unexplored. Here we performed single-cell RNA sequencing on 20 organs to reveal cell-type-specific responses to young and aged blood in heterochronic parabiosis. Adipose mesenchymal stromal cells, haematopoietic stem cells and hepatocytes are among those cell types that are especially responsive. On the pathway level, young blood invokes new gene sets in addition to reversing established ageing patterns, with the global rescue of genes encoding electron transport chain subunits pinpointing a prominent role of mitochondrial function in parabiosis-mediated rejuvenation. We observed an almost universal loss of gene expression with age that is largely mimicked by parabiosis: aged blood reduces global gene expression, and young blood restores it in select cell types. Together, these data lay the groundwork for a systemic understanding of the interplay between blood-borne factors and cellular integrity.


Asunto(s)
Parabiosis , Análisis de la Célula Individual , Adipocitos , Envejecimiento/genética , Transporte de Electrón/genética , Células Madre Hematopoyéticas , Hepatocitos , Células Madre Mesenquimatosas , Mitocondrias , Especificidad de Órganos/genética , RNA-Seq , Rejuvenecimiento
6.
Nature ; 602(7898): 689-694, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-35140405

RESUMEN

Liquid biopsies that measure circulating cell-free RNA (cfRNA) offer an opportunity to study the development of pregnancy-related complications in a non-invasive manner and to bridge gaps in clinical care1-4. Here we used 404 blood samples from 199 pregnant mothers to identify and validate cfRNA transcriptomic changes that are associated with preeclampsia, a multi-organ syndrome that is the second largest cause of maternal death globally5. We find that changes in cfRNA gene expression between normotensive and preeclamptic mothers are marked and stable early in gestation, well before the onset of symptoms. These changes are enriched for genes specific to neuromuscular, endothelial and immune cell types and tissues that reflect key aspects of preeclampsia physiology6-9, suggest new hypotheses for disease progression and correlate with maternal organ health. This enabled the identification and independent validation of a panel of 18 genes that when measured between 5 and 16 weeks of gestation can form the basis of a liquid biopsy test that would identify mothers at risk of preeclampsia long before clinical symptoms manifest themselves. Tests based on these observations could help predict and manage who is at risk for preeclampsia-an important objective for obstetric care10,11.


Asunto(s)
Ácidos Nucleicos Libres de Células , Diagnóstico Precoz , Preeclampsia , ARN , Presión Sanguínea , Ácidos Nucleicos Libres de Células/sangre , Ácidos Nucleicos Libres de Células/genética , Femenino , Humanos , Madres , Preeclampsia/diagnóstico , Preeclampsia/genética , Embarazo , ARN/sangre , ARN/genética , Transcriptoma
7.
Nature ; 597(7875): 256-262, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34381212

RESUMEN

Loss of skeletal integrity during ageing and disease is associated with an imbalance in the opposing actions of osteoblasts and osteoclasts1. Here we show that intrinsic ageing of skeletal stem cells (SSCs)2 in mice alters signalling in the bone marrow niche and skews the differentiation of bone and blood lineages, leading to fragile bones that regenerate poorly. Functionally, aged SSCs have a decreased bone- and cartilage-forming potential but produce more stromal lineages that express high levels of pro-inflammatory and pro-resorptive cytokines. Single-cell RNA-sequencing studies link the functional loss to a diminished transcriptomic diversity of SSCs in aged mice, which thereby contributes to the transformation of the bone marrow niche. Exposure to a youthful circulation through heterochronic parabiosis or systemic reconstitution with young haematopoietic stem cells did not reverse the diminished osteochondrogenic activity of aged SSCs, or improve bone mass or skeletal healing parameters in aged mice. Conversely, the aged SSC lineage promoted osteoclastic activity and myeloid skewing by haematopoietic stem and progenitor cells, suggesting that the ageing of SSCs is a driver of haematopoietic ageing. Deficient bone regeneration in aged mice could only be returned to youthful levels by applying a combinatorial treatment of BMP2 and a CSF1 antagonist locally to fractures, which reactivated aged SSCs and simultaneously ablated the inflammatory, pro-osteoclastic milieu. Our findings provide mechanistic insights into the complex, multifactorial mechanisms that underlie skeletal ageing and offer prospects for rejuvenating the aged skeletal system.


Asunto(s)
Envejecimiento/patología , Huesos/patología , Senescencia Celular , Inflamación/patología , Nicho de Células Madre , Células Madre/patología , Animales , Proteína Morfogenética Ósea 2/metabolismo , Regeneración Ósea , Linaje de la Célula , Femenino , Curación de Fractura , Hematopoyesis , Factor Estimulante de Colonias de Macrófagos/metabolismo , Masculino , Ratones , Células Mieloides/citología , Osteoclastos/citología , Rejuvenecimiento
8.
Nature ; 583(7817): 596-602, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32669715

RESUMEN

Ageing is the single greatest cause of disease and death worldwide, and understanding the associated processes could vastly improve quality of life. Although major categories of ageing damage have been identified-such as altered intercellular communication, loss of proteostasis and eroded mitochondrial function1-these deleterious processes interact with extraordinary complexity within and between organs, and a comprehensive, whole-organism analysis of ageing dynamics has been lacking. Here we performed bulk RNA sequencing of 17 organs and plasma proteomics at 10 ages across the lifespan of Mus musculus, and integrated these findings with data from the accompanying Tabula Muris Senis2-or 'Mouse Ageing Cell Atlas'-which follows on from the original Tabula Muris3. We reveal linear and nonlinear shifts in gene expression during ageing, with the associated genes clustered in consistent trajectory groups with coherent biological functions-including extracellular matrix regulation, unfolded protein binding, mitochondrial function, and inflammatory and immune response. Notably, these gene sets show similar expression across tissues, differing only in the amplitude and the age of onset of expression. Widespread activation of immune cells is especially pronounced, and is first detectable in white adipose depots during middle age. Single-cell RNA sequencing confirms the accumulation of T cells and B cells in adipose tissue-including plasma cells that express immunoglobulin J-which also accrue concurrently across diverse organs. Finally, we show how gene expression shifts in distinct tissues are highly correlated with corresponding protein levels in plasma, thus potentially contributing to the ageing of the systemic circulation. Together, these data demonstrate a similar yet asynchronous inter- and intra-organ progression of ageing, providing a foundation from which to track systemic sources of declining health at old age.


Asunto(s)
Envejecimiento/genética , Envejecimiento/fisiología , Regulación de la Expresión Génica , Especificidad de Órganos/genética , Animales , Proteínas Sanguíneas/análisis , Proteínas Sanguíneas/genética , Femenino , Cadenas J de Inmunoglobulina/genética , Cadenas J de Inmunoglobulina/metabolismo , Masculino , Ratones , Células Plasmáticas/citología , Células Plasmáticas/metabolismo , ARN Mensajero/análisis , ARN Mensajero/genética , RNA-Seq , Análisis de la Célula Individual , Linfocitos T/citología , Linfocitos T/metabolismo , Factores de Tiempo , Transcriptoma
9.
Nature ; 564(7736): 425-429, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30518860

RESUMEN

Haematopoiesis is an essential process that evolved in multicellular animals. At the heart of this process are haematopoietic stem cells (HSCs), which are multipotent and self-renewing, and generate the entire repertoire of blood and immune cells throughout an animal's life1. Although there have been comprehensive studies on self-renewal, differentiation, physiological regulation and niche occupation in vertebrate HSCs, relatively little is known about the evolutionary origin and niches of these cells. Here we describe the haematopoietic system of Botryllus schlosseri, a colonial tunicate that has a vasculature and circulating blood cells, and interesting stem-cell biology and immunity characteristics2-8. Self-recognition between genetically compatible B. schlosseri colonies leads to the formation of natural parabionts with shared circulation, whereas incompatible colonies reject each other3,4,7. Using flow cytometry, whole-transcriptome sequencing of defined cell populations and diverse functional assays, we identify HSCs, progenitors, immune effector cells and an HSC niche, and demonstrate that self-recognition inhibits allospecific cytotoxic reactions. Our results show that HSC and myeloid lineage immune cells emerged in a common ancestor of tunicates and vertebrates, and also suggest that haematopoietic bone marrow and the B. schlosseri endostyle niche evolved from a common origin.


Asunto(s)
Hematopoyesis , Sistema Hematopoyético/citología , Mamíferos/sangre , Filogenia , Urocordados/citología , Animales , Diferenciación Celular , Linaje de la Célula , Citotoxicidad Inmunológica , Femenino , Citometría de Flujo , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/inmunología , Inmunidad Celular , Isoantígenos/inmunología , Masculino , Mamíferos/anatomía & histología , Células Mieloides/citología , Células Mieloides/inmunología , Fagocitosis/inmunología , Nicho de Células Madre , Transcriptoma/genética , Urocordados/anatomía & histología , Urocordados/genética , Urocordados/inmunología
10.
Nature ; 534(7607): 391-5, 2016 06 16.
Artículo en Inglés | MEDLINE | ID: mdl-27281220

RESUMEN

Direct lineage reprogramming represents a remarkable conversion of cellular and transcriptome states. However, the intermediate stages through which individual cells progress during reprogramming are largely undefined. Here we use single-cell RNA sequencing at multiple time points to dissect direct reprogramming from mouse embryonic fibroblasts to induced neuronal cells. By deconstructing heterogeneity at each time point and ordering cells by transcriptome similarity, we find that the molecular reprogramming path is remarkably continuous. Overexpression of the proneural pioneer factor Ascl1 results in a well-defined initialization, causing cells to exit the cell cycle and re-focus gene expression through distinct neural transcription factors. The initial transcriptional response is relatively homogeneous among fibroblasts, suggesting that the early steps are not limiting for productive reprogramming. Instead, the later emergence of a competing myogenic program and variable transgene dynamics over time appear to be the major efficiency limits of direct reprogramming. Moreover, a transcriptional state, distinct from donor and target cell programs, is transiently induced in cells undergoing productive reprogramming. Our data provide a high-resolution approach for understanding transcriptome states during lineage differentiation.


Asunto(s)
Reprogramación Celular/genética , Fibroblastos/citología , Fibroblastos/metabolismo , Neuronas/citología , Neuronas/metabolismo , Análisis de Secuencia de ARN , Análisis de la Célula Individual , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Ciclo Celular/genética , Linaje de la Célula/genética , Transdiferenciación Celular/genética , Embrión de Mamíferos/citología , Perfilación de la Expresión Génica , Silenciador del Gen , Proteínas de Homeodominio/metabolismo , Ratones , Proteínas del Tejido Nervioso/metabolismo , Factores del Dominio POU/metabolismo , Factores de Tiempo , Factores de Transcripción/metabolismo , Transcriptoma/genética , Transgenes/genética
11.
Nucleic Acids Res ; 47(14): e83, 2019 08 22.
Artículo en Inglés | MEDLINE | ID: mdl-31114866

RESUMEN

The growing prevalence of deadly microbes with resistance to previously life-saving drug therapies is a dire threat to human health. Detection of low abundance pathogen sequences remains a challenge for metagenomic Next Generation Sequencing (NGS). We introduce FLASH (Finding Low Abundance Sequences by Hybridization), a next-generation CRISPR/Cas9 diagnostic method that takes advantage of the efficiency, specificity and flexibility of Cas9 to enrich for a programmed set of sequences. FLASH-NGS achieves up to 5 orders of magnitude of enrichment and sub-attomolar gene detection with minimal background. We provide an open-source software tool (FLASHit) for guide RNA design. Here we applied it to detection of antimicrobial resistance genes in respiratory fluid and dried blood spots, but FLASH-NGS is applicable to all areas that rely on multiplex PCR.


Asunto(s)
Antibacterianos/farmacología , Sistemas CRISPR-Cas , Biología Computacional/métodos , Farmacorresistencia Bacteriana/efectos de los fármacos , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Bacterias/clasificación , Bacterias/efectos de los fármacos , Bacterias/genética , Infecciones Bacterianas/diagnóstico , Infecciones Bacterianas/genética , Infecciones Bacterianas/prevención & control , Farmacorresistencia Bacteriana/genética , Humanos , Metagenómica/métodos , Reproducibilidad de los Resultados , Sensibilidad y Especificidad
12.
Nature ; 509(7500): 371-5, 2014 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-24739965

RESUMEN

The mammalian lung is a highly branched network in which the distal regions of the bronchial tree transform during development into a densely packed honeycomb of alveolar air sacs that mediate gas exchange. Although this transformation has been studied by marker expression analysis and fate-mapping, the mechanisms that control the progression of lung progenitors along distinct lineages into mature alveolar cell types are still incompletely known, in part because of the limited number of lineage markers and the effects of ensemble averaging in conventional transcriptome analysis experiments on cell populations. Here we show that single-cell transcriptome analysis circumvents these problems and enables direct measurement of the various cell types and hierarchies in the developing lung. We used microfluidic single-cell RNA sequencing (RNA-seq) on 198 individual cells at four different stages encompassing alveolar differentiation to measure the transcriptional states which define the developmental and cellular hierarchy of the distal mouse lung epithelium. We empirically classified cells into distinct groups by using an unbiased genome-wide approach that did not require a priori knowledge of the underlying cell types or the previous purification of cell populations. The results confirmed the basic outlines of the classical model of epithelial cell-type diversity in the distal lung and led to the discovery of many previously unknown cell-type markers, including transcriptional regulators that discriminate between the different populations. We reconstructed the molecular steps during maturation of bipotential progenitors along both alveolar lineages and elucidated the full life cycle of the alveolar type 2 cell lineage. This single-cell genomics approach is applicable to any developing or mature tissue to robustly delineate molecularly distinct cell types, define progenitors and lineage hierarchies, and identify lineage-specific regulatory factors.


Asunto(s)
Linaje de la Célula/genética , Células Epiteliales/citología , Células Epiteliales/metabolismo , Pulmón/citología , Análisis de Secuencia de ARN/métodos , Análisis de la Célula Individual/métodos , Animales , Bronquios/citología , Diferenciación Celular/genética , Células Epiteliales/clasificación , Femenino , Marcadores Genéticos , Genoma/genética , Genómica , Pulmón/embriología , Ratones , Ratones Endogámicos C57BL , Alveolos Pulmonares/citología , Intercambio Gaseoso Pulmonar , Células Madre/citología , Transcriptoma/genética
13.
Proc Natl Acad Sci U S A ; 114(36): 9623-9628, 2017 09 05.
Artículo en Inglés | MEDLINE | ID: mdl-28830999

RESUMEN

Blood circulates throughout the human body and contains molecules drawn from virtually every tissue, including the microbes and viruses which colonize the body. Through massive shotgun sequencing of circulating cell-free DNA from the blood, we identified hundreds of new bacteria and viruses which represent previously unidentified members of the human microbiome. Analyzing cumulative sequence data from 1,351 blood samples collected from 188 patients enabled us to assemble 7,190 contiguous regions (contigs) larger than 1 kbp, of which 3,761 are novel with little or no sequence homology in any existing databases. The vast majority of these novel contigs possess coding sequences, and we have validated their existence both by finding their presence in independent experiments and by performing direct PCR amplification. When their nearest neighbors are located in the tree of life, many of the organisms represent entirely novel taxa, showing that microbial diversity within the human body is substantially broader than previously appreciated.


Asunto(s)
Ácidos Nucleicos Libres de Células/sangre , Ácidos Nucleicos Libres de Células/genética , ADN Bacteriano/sangre , ADN Bacteriano/genética , ADN Viral/sangre , ADN Viral/genética , Microbiota/genética , Variación Genética , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Humanos , Metagenómica/métodos , Filogenia
14.
Proc Natl Acad Sci U S A ; 113(41): 11555-11560, 2016 10 11.
Artículo en Inglés | MEDLINE | ID: mdl-27681629

RESUMEN

Somatic mosaicism, the occurrence and propagation of genetic variation in cell lineages after fertilization, is increasingly recognized to play a causal role in a variety of human diseases. We investigated the case of life-threatening arrhythmia in a 10-day-old infant with long QT syndrome (LQTS). Rapid genome sequencing suggested a variant in the sodium channel NaV1.5 encoded by SCN5A, NM_000335:c.5284G > T predicting p.(V1762L), but read depth was insufficient to be diagnostic. Exome sequencing of the trio confirmed read ratios inconsistent with Mendelian inheritance only in the proband. Genotyping of single circulating leukocytes demonstrated the mutation in the genomes of 8% of patient cells, and RNA sequencing of cardiac tissue from the infant confirmed the expression of the mutant allele at mosaic ratios. Heterologous expression of the mutant channel revealed significantly delayed sodium current with a dominant negative effect. To investigate the mechanism by which mosaicism might cause arrhythmia, we built a finite element simulation model incorporating Purkinje fiber activation. This model confirmed the pathogenic consequences of cardiac cellular mosaicism and, under the presenting conditions of this case, recapitulated 2:1 AV block and arrhythmia. To investigate the extent to which mosaicism might explain undiagnosed arrhythmia, we studied 7,500 affected probands undergoing commercial gene-panel testing. Four individuals with pathogenic variants arising from early somatic mutation events were found. Here we establish cardiac mosaicism as a causal mechanism for LQTS and present methods by which the general phenomenon, likely to be relevant for all genetic diseases, can be detected through single-cell analysis and next-generation sequencing.


Asunto(s)
Predisposición Genética a la Enfermedad , Síndrome de QT Prolongado/genética , Mosaicismo , Potenciales de Acción , Arritmias Cardíacas/complicaciones , Arritmias Cardíacas/genética , Arritmias Cardíacas/fisiopatología , Secuencia de Bases , Cardiomiopatía Dilatada/complicaciones , Cardiomiopatía Dilatada/genética , Cardiomiopatía Dilatada/fisiopatología , Simulación por Computador , Difusión , Electrocardiografía , Frecuencia de los Genes/genética , Genes Dominantes , Sitios Genéticos , Técnicas de Genotipaje , Sistema de Conducción Cardíaco/fisiopatología , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Lactante , Activación del Canal Iónico/genética , Síndrome de QT Prolongado/complicaciones , Síndrome de QT Prolongado/diagnóstico por imagen , Síndrome de QT Prolongado/fisiopatología , Modelos Biológicos , Mutación/genética , Miocitos Cardíacos/metabolismo , Canal de Sodio Activado por Voltaje NAV1.5/genética , Fenotipo , Análisis de la Célula Individual
15.
Proc Natl Acad Sci U S A ; 112(43): 13336-41, 2015 Oct 27.
Artículo en Inglés | MEDLINE | ID: mdl-26460048

RESUMEN

The survival rate following lung transplantation is among the lowest of all solid-organ transplants, and current diagnostic tests often fail to distinguish between infection and rejection, the two primary posttransplant clinical complications. We describe a diagnostic assay that simultaneously monitors for rejection and infection in lung transplant recipients by sequencing of cell-free DNA (cfDNA) in plasma. We determined that the levels of donor-derived cfDNA directly correlate with the results of invasive tests of rejection (area under the curve 0.9). We also analyzed the nonhuman cfDNA as a hypothesis-free approach to test for infections. Cytomegalovirus is most frequently assayed clinically, and the levels of CMV-derived sequences in cfDNA are consistent with clinical results. We furthermore show that hypothesis-free monitoring for pathogens using cfDNA reveals undiagnosed cases of infection, and that certain infectious pathogens such as human herpesvirus (HHV) 6, HHV-7, and adenovirus, which are not often tested clinically, occur with high frequency in this cohort.


Asunto(s)
ADN Viral/sangre , Rechazo de Injerto/diagnóstico , Trasplante de Pulmón/efectos adversos , Cuidados Posoperatorios/métodos , Infección de la Herida Quirúrgica/diagnóstico , Secuencia de Bases , Citomegalovirus/genética , Humanos , Datos de Secuencia Molecular , Polimorfismo de Nucleótido Simple/genética , Análisis de Secuencia de ADN , Especificidad de la Especie , Infección de la Herida Quirúrgica/virología
16.
Nat Methods ; 11(1): 41-6, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24141493

RESUMEN

Interest in single-cell whole-transcriptome analysis is growing rapidly, especially for profiling rare or heterogeneous populations of cells. We compared commercially available single-cell RNA amplification methods with both microliter and nanoliter volumes, using sequence from bulk total RNA and multiplexed quantitative PCR as benchmarks to systematically evaluate the sensitivity and accuracy of various single-cell RNA-seq approaches. We show that single-cell RNA-seq can be used to perform accurate quantitative transcriptome measurement in individual cells with a relatively small number of sequencing reads and that sequencing large numbers of single cells can recapitulate bulk transcriptome complexity.


Asunto(s)
Análisis de Secuencia de ARN/métodos , Análisis de la Célula Individual/métodos , Interpretación Estadística de Datos , Procesamiento Automatizado de Datos , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Células HCT116 , Humanos , Microfluídica , Técnicas de Amplificación de Ácido Nucleico , Reacción en Cadena de la Polimerasa/métodos , Reproducibilidad de los Resultados , Análisis de Secuencia de ADN , Transcriptoma
17.
Blood Adv ; 8(7): 1820-1833, 2024 Apr 09.
Artículo en Inglés | MEDLINE | ID: mdl-38096800

RESUMEN

ABSTRACT: Recombination-activating genes (RAG1 and RAG2) are critical for lymphoid cell development and function by initiating the variable (V), diversity (D), and joining (J) (V(D)J)-recombination process to generate polyclonal lymphocytes with broad antigen specificity. The clinical manifestations of defective RAG1/2 genes range from immune dysregulation to severe combined immunodeficiencies (SCIDs), causing life-threatening infections and death early in life without hematopoietic cell transplantation (HCT). Despite improvements, haploidentical HCT without myeloablative conditioning carries a high risk of graft failure and incomplete immune reconstitution. The RAG complex is only expressed during the G0-G1 phase of the cell cycle in the early stages of T- and B-cell development, underscoring that a direct gene correction might capture the precise temporal expression of the endogenous gene. Here, we report a feasibility study using the CRISPR/Cas9-based "universal gene-correction" approach for the RAG2 locus in human hematopoietic stem/progenitor cells (HSPCs) from healthy donors and RAG2-SCID patient. V(D)J-recombinase activity was restored after gene correction of RAG2-SCID-derived HSPCs, resulting in the development of T-cell receptor (TCR) αß and γδ CD3+ cells and single-positive CD4+ and CD8+ lymphocytes. TCR repertoire analysis indicated a normal distribution of CDR3 length and preserved usage of the distal TRAV genes. We confirmed the in vivo rescue of B-cell development with normal immunoglobulin M surface expression and a significant decrease in CD56bright natural killer cells. Together, we provide specificity, toxicity, and efficacy data supporting the development of a gene-correction therapy to benefit RAG2-deficient patients.


Asunto(s)
Proteínas de Homeodominio , Inmunodeficiencia Combinada Grave , Humanos , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Células Madre Hematopoyéticas/metabolismo , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Proteínas Nucleares , Receptores de Antígenos de Linfocitos T alfa-beta/genética , Inmunodeficiencia Combinada Grave/genética , Inmunodeficiencia Combinada Grave/terapia , VDJ Recombinasas
18.
Elife ; 122023 01 31.
Artículo en Inglés | MEDLINE | ID: mdl-36719070

RESUMEN

Nutrient availability fluctuates in most natural populations, forcing organisms to undergo periods of fasting and re-feeding. It is unknown how dietary changes influence liver homeostasis. Here, we show that a switch from ad libitum feeding to intermittent fasting (IF) promotes rapid hepatocyte proliferation. Mechanistically, IF-induced hepatocyte proliferation is driven by the combined action of systemic FGF15 and localized WNT signaling. Hepatocyte proliferation during periods of fasting and re-feeding re-establishes a constant liver-to-body mass ratio, thus maintaining the hepatostat. This study provides the first example of dietary influence on adult hepatocyte proliferation and challenges the widely held view that liver tissue is mostly quiescent unless chemically or mechanically injured.


Asunto(s)
Ayuno Intermitente , Regeneración Hepática , Ratones , Animales , Hígado , Ayuno , Hepatocitos , Proliferación Celular
19.
Life Sci Alliance ; 6(1)2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36526371

RESUMEN

Spatial transcriptomics extends single-cell RNA sequencing (scRNA-seq) by providing spatial context for cell type identification and analysis. Imaging-based spatial technologies such as multiplexed error-robust fluorescence in situ hybridization (MERFISH) can achieve single-cell resolution, directly mapping single-cell identities to spatial positions. MERFISH produces a different data type than scRNA-seq, and a technical comparison between the two modalities is necessary to ascertain how to best integrate them. We performed MERFISH on the mouse liver and kidney and compared the resulting bulk and single-cell RNA statistics with those from the Tabula Muris Senis cell atlas and from two Visium datasets. MERFISH quantitatively reproduced the bulk RNA-seq and scRNA-seq results with improvements in overall dropout rates and sensitivity. Finally, we found that MERFISH independently resolved distinct cell types and spatial structure in both the liver and kidney. Computational integration with the Tabula Muris Senis atlas did not enhance these results. We conclude that MERFISH provides a quantitatively comparable method for single-cell gene expression and can identify cell types without the need for computational integration with scRNA-seq atlases.


Asunto(s)
Análisis de la Célula Individual , Transcriptoma , Ratones , Animales , Hibridación Fluorescente in Situ/métodos , Análisis de la Célula Individual/métodos , Transcriptoma/genética , Perfilación de la Expresión Génica/métodos , RNA-Seq
20.
bioRxiv ; 2023 Oct 30.
Artículo en Inglés | MEDLINE | ID: mdl-37808827

RESUMEN

Humans constantly encounter new microbes, but few become long-term residents of the adult gut microbiome. Classical theories predict that colonization is determined by the availability of open niches, but it remains unclear whether other ecological barriers limit commensal colonization in natural settings. To disentangle these effects, we used a controlled perturbation with the antibiotic ciprofloxacin to investigate the dynamics of gut microbiome transmission in 22 households of healthy, cohabiting adults. Colonization was rare in three-quarters of antibiotic-taking subjects, whose resident strains rapidly recovered in the week after antibiotics ended. In contrast, the remaining antibiotic-taking subjects exhibited lasting responses, with extensive species losses and transient expansions of potential opportunistic pathogens. These subjects experienced elevated rates of commensal colonization, but only after long delays: many new colonizers underwent sudden, correlated expansions months after the antibiotic perturbation. Furthermore, strains that had previously transmitted between cohabiting partners rarely recolonized after antibiotic disruptions, showing that colonization displays substantial historical contingency. This work demonstrates that there remain substantial ecological barriers to colonization even after major microbiome disruptions, suggesting that dispersal interactions and priority effects limit the pace of community change.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA