Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 78
Filtrar
1.
Cell ; 187(13): 3236-3248.e21, 2024 Jun 20.
Artículo en Inglés | MEDLINE | ID: mdl-38772369

RESUMEN

Leveraging AAVs' versatile tropism and labeling capacity, we expanded the scale of in vivo CRISPR screening with single-cell transcriptomic phenotyping across embryonic to adult brains and peripheral nervous systems. Through extensive tests of 86 vectors across AAV serotypes combined with a transposon system, we substantially amplified labeling efficacy and accelerated in vivo gene delivery from weeks to days. Our proof-of-principle in utero screen identified the pleiotropic effects of Foxg1, highlighting its tight regulation of distinct networks essential for cell fate specification of Layer 6 corticothalamic neurons. Notably, our platform can label >6% of cerebral cells, surpassing the current state-of-the-art efficacy at <0.1% by lentivirus, to achieve analysis of over 30,000 cells in one experiment and enable massively parallel in vivo Perturb-seq. Compatible with various phenotypic measurements (single-cell or spatial multi-omics), it presents a flexible approach to interrogate gene function across cell types in vivo, translating gene variants to their causal function.


Asunto(s)
Redes Reguladoras de Genes , Análisis de la Célula Individual , Animales , Femenino , Humanos , Ratones , Corteza Cerebral/metabolismo , Corteza Cerebral/citología , Sistemas CRISPR-Cas/genética , Dependovirus/genética , Factores de Transcripción Forkhead/metabolismo , Factores de Transcripción Forkhead/genética , Vectores Genéticos/metabolismo , Ratones Endogámicos C57BL , Proteínas del Tejido Nervioso/metabolismo , Proteínas del Tejido Nervioso/genética , Neuronas/metabolismo , Neuronas/citología , Análisis de la Célula Individual/métodos , Transcriptoma/genética , Línea Celular , Transcripción Genética
2.
Cell ; 185(20): 3770-3788.e27, 2022 09 29.
Artículo en Inglés | MEDLINE | ID: mdl-36179669

RESUMEN

Realizing the full utility of brain organoids to study human development requires understanding whether organoids precisely replicate endogenous cellular and molecular events, particularly since acquisition of cell identity in organoids can be impaired by abnormal metabolic states. We present a comprehensive single-cell transcriptomic, epigenetic, and spatial atlas of human cortical organoid development, comprising over 610,000 cells, from generation of neural progenitors through production of differentiated neuronal and glial subtypes. We show that processes of cellular diversification correlate closely to endogenous ones, irrespective of metabolic state, empowering the use of this atlas to study human fate specification. We define longitudinal molecular trajectories of cortical cell types during organoid development, identify genes with predicted human-specific roles in lineage establishment, and uncover early transcriptional diversity of human callosal neurons. The findings validate this comprehensive atlas of human corticogenesis in vitro as a resource to prime investigation into the mechanisms of human cortical development.


Asunto(s)
Corteza Cerebral , Organoides , Diferenciación Celular , Corteza Cerebral/metabolismo , Humanos , Neurogénesis , Neuronas , Organoides/metabolismo
3.
Nat Immunol ; 25(4): 671-681, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38448779

RESUMEN

Cognitive impairment is a frequent manifestation of neuropsychiatric systemic lupus erythematosus, present in up to 80% of patients and leading to a diminished quality of life. In the present study, we used a model of lupus-like cognitive impairment that is initiated when antibodies that crossreact with excitatory neuronal receptors penetrate the hippocampus, causing immediate, self-limited, excitotoxic death of hippocampal neurons, which is then followed by a significant loss of dendritic complexity in surviving neurons. This injury creates a maladaptive equilibrium that is sustained in mice for at least 1 year. We identified a feedforward loop of microglial activation and microglia-dependent synapse elimination dependent on neuronal secretion of high mobility group box 1 protein (HMGB1) which binds the receptor for advanced glycation end products (RAGE) and leads to microglial secretion of C1q, upregulation of interleukin-10 with consequent downregulation of leukocyte-associated immunoglobulin-like receptor 1 (LAIR-1), an inhibitory receptor for C1q. Treatment with a centrally acting angiotensin-converting enzyme inhibitor or with an angiotensin-receptor blocker restored a healthy equilibrium, microglial quiescence and intact spatial memory.


Asunto(s)
Autoanticuerpos , Proteína HMGB1 , Animales , Ratones , Complemento C1q , Proteína HMGB1/metabolismo , Enfermedades Neuroinflamatorias , Calidad de Vida , Receptor para Productos Finales de Glicación Avanzada/metabolismo
4.
Cell ; 174(6): 1477-1491.e19, 2018 09 06.
Artículo en Inglés | MEDLINE | ID: mdl-30146158

RESUMEN

Aging is a major risk factor for both genetic and sporadic neurodegenerative disorders. However, it is unclear how aging interacts with genetic predispositions to promote neurodegeneration. Here, we investigate how partial loss of function of TBK1, a major genetic cause for amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD) comorbidity, leads to age-dependent neurodegeneration. We show that TBK1 is an endogenous inhibitor of RIPK1 and the embryonic lethality of Tbk1-/- mice is dependent on RIPK1 kinase activity. In aging human brains, another endogenous RIPK1 inhibitor, TAK1, exhibits a marked decrease in expression. We show that in Tbk1+/- mice, the reduced myeloid TAK1 expression promotes all the key hallmarks of ALS/FTD, including neuroinflammation, TDP-43 aggregation, axonal degeneration, neuronal loss, and behavior deficits, which are blocked upon inhibition of RIPK1. Thus, aging facilitates RIPK1 activation by reducing TAK1 expression, which cooperates with genetic risk factors to promote the onset of ALS/FTD.


Asunto(s)
Apoptosis , Proteínas Serina-Treonina Quinasas/metabolismo , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Adulto , Anciano , Envejecimiento , Animales , Apoptosis/efectos de los fármacos , Axones/metabolismo , Conducta Animal , Encéfalo/citología , Encéfalo/metabolismo , Células Cultivadas , Humanos , Quinasa I-kappa B/metabolismo , Ratones , Ratones Noqueados , Microglía/citología , Microglía/efectos de los fármacos , Microglía/metabolismo , Fosforilación/efectos de los fármacos , Proteínas Serina-Treonina Quinasas/deficiencia , Proteínas Serina-Treonina Quinasas/genética , Proteína Serina-Treonina Quinasas de Interacción con Receptores/deficiencia , Proteína Serina-Treonina Quinasas de Interacción con Receptores/genética , Médula Espinal/metabolismo , Estaurosporina/farmacología , Factor de Necrosis Tumoral alfa/farmacología
5.
Cell ; 166(5): 1308-1323.e30, 2016 Aug 25.
Artículo en Inglés | MEDLINE | ID: mdl-27565351

RESUMEN

Patterns of gene expression can be used to characterize and classify neuronal types. It is challenging, however, to generate taxonomies that fulfill the essential criteria of being comprehensive, harmonizing with conventional classification schemes, and lacking superfluous subdivisions of genuine types. To address these challenges, we used massively parallel single-cell RNA profiling and optimized computational methods on a heterogeneous class of neurons, mouse retinal bipolar cells (BCs). From a population of ∼25,000 BCs, we derived a molecular classification that identified 15 types, including all types observed previously and two novel types, one of which has a non-canonical morphology and position. We validated the classification scheme and identified dozens of novel markers using methods that match molecular expression to cell morphology. This work provides a systematic methodology for achieving comprehensive molecular classification of neurons, identifies novel neuronal types, and uncovers transcriptional differences that distinguish types within a class.


Asunto(s)
Células Bipolares de la Retina/clasificación , Transcriptoma , Células Amacrinas/citología , Animales , Análisis por Conglomerados , Femenino , Marcadores Genéticos , Masculino , Ratones , Ratones Endogámicos , Ratones Transgénicos , Células Bipolares de la Retina/citología , Células Bipolares de la Retina/metabolismo , Análisis de Secuencia de ARN , Análisis de la Célula Individual/métodos , Transcripción Genética
6.
Nature ; 631(8019): 142-149, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38926573

RESUMEN

Interindividual genetic variation affects the susceptibility to and progression of many diseases1,2. However, efforts to study how individual human brains differ in normal development and disease phenotypes are limited by the paucity of faithful cellular human models, and the difficulty of scaling current systems to represent multiple people. Here we present human brain Chimeroids, a highly reproducible, multidonor human brain cortical organoid model generated by the co-development of cells from a panel of individual donors in a single organoid. By reaggregating cells from multiple single-donor organoids at the neural stem cell or neural progenitor cell stage, we generate Chimeroids in which each donor produces all cell lineages of the cerebral cortex, even when using pluripotent stem cell lines with notable growth biases. We used Chimeroids to investigate interindividual variation in the susceptibility to neurotoxic triggers that exhibit high clinical phenotypic variability: ethanol and the antiepileptic drug valproic acid. Individual donors varied in both the penetrance of the effect on target cell types, and the molecular phenotype within each affected cell type. Our results suggest that human genetic background may be an important mediator of neurotoxin susceptibility and introduce Chimeroids as a scalable system for high-throughput investigation of interindividual variation in processes of brain development and disease.


Asunto(s)
Células-Madre Neurales , Organoides , Humanos , Organoides/efectos de los fármacos , Organoides/citología , Células-Madre Neurales/efectos de los fármacos , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Masculino , Linaje de la Célula/efectos de los fármacos , Encéfalo/efectos de los fármacos , Encéfalo/citología , Corteza Cerebral/efectos de los fármacos , Corteza Cerebral/citología , Neurotoxinas/toxicidad , Fenotipo , Femenino , Susceptibilidad a Enfermedades , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/efectos de los fármacos , Células Madre Pluripotentes/metabolismo , Donantes de Tejidos , Línea Celular
7.
Nature ; 608(7924): 750-756, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35948630

RESUMEN

Microglia are specialized macrophages in the brain parenchyma that exist in multiple transcriptional states and reside within a wide range of neuronal environments1-4. However, how and where these states are generated remains poorly understood. Here, using the mouse somatosensory cortex, we demonstrate that microglia density and molecular state acquisition are determined by the local composition of pyramidal neuron classes. Using single-cell and spatial transcriptomic profiling, we unveil the molecular signatures and spatial distributions of diverse microglia populations and show that certain states are enriched in specific cortical layers, whereas others are broadly distributed throughout the cortex. Notably, conversion of deep-layer pyramidal neurons to an alternate class identity reconfigures the distribution of local, layer-enriched homeostatic microglia to match the new neuronal niche. Leveraging the transcriptional diversity of pyramidal neurons in the neocortex, we construct a ligand-receptor atlas describing interactions between individual pyramidal neuron subtypes and microglia states, revealing rules of neuron-microglia communication. Our findings uncover a fundamental role for neuronal diversity in instructing the acquisition of microglia states as a potential mechanism for fine-tuning neuroimmune interactions within the cortical local circuitry.


Asunto(s)
Microglía , Neocórtex , Células Piramidales , Corteza Somatosensorial , Animales , Recuento de Células , Ratones , Microglía/clasificación , Microglía/fisiología , Neocórtex/citología , Neocórtex/fisiología , Células Piramidales/clasificación , Células Piramidales/fisiología , Análisis de la Célula Individual , Corteza Somatosensorial/citología , Corteza Somatosensorial/fisiología , Transcriptoma
8.
Nature ; 603(7901): 455-463, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35264797

RESUMEN

Mutations in non-coding regulatory DNA sequences can alter gene expression, organismal phenotype and fitness1-3. Constructing complete fitness landscapes, in which DNA sequences are mapped to fitness, is a long-standing goal in biology, but has remained elusive because it is challenging to generalize reliably to vast sequence spaces4-6. Here we build sequence-to-expression models that capture fitness landscapes and use them to decipher principles of regulatory evolution. Using millions of randomly sampled promoter DNA sequences and their measured expression levels in the yeast Saccharomyces cerevisiae, we learn deep neural network models that generalize with excellent prediction performance, and enable sequence design for expression engineering. Using our models, we study expression divergence under genetic drift and strong-selection weak-mutation regimes to find that regulatory evolution is rapid and subject to diminishing returns epistasis; that conflicting expression objectives in different environments constrain expression adaptation; and that stabilizing selection on gene expression leads to the moderation of regulatory complexity. We present an approach for using such models to detect signatures of selection on expression from natural variation in regulatory sequences and use it to discover an instance of convergent regulatory evolution. We assess mutational robustness, finding that regulatory mutation effect sizes follow a power law, characterize regulatory evolvability, visualize promoter fitness landscapes, discover evolvability archetypes and illustrate the mutational robustness of natural regulatory sequence populations. Our work provides a general framework for designing regulatory sequences and addressing fundamental questions in regulatory evolution.


Asunto(s)
Flujo Genético , Modelos Genéticos , Evolución Biológica , ADN , Evolución Molecular , Regulación de la Expresión Génica , Mutación/genética , Fenotipo , Saccharomyces cerevisiae/genética
9.
Nature ; 602(7896): 268-273, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-35110736

RESUMEN

Genetic risk for autism spectrum disorder (ASD) is associated with hundreds of genes spanning a wide range of biological functions1-6. The alterations in the human brain resulting from mutations in these genes remain unclear. Furthermore, their phenotypic manifestation varies across individuals7,8. Here we used organoid models of the human cerebral cortex to identify cell-type-specific developmental abnormalities that result from haploinsufficiency in three ASD risk genes-SUV420H1 (also known as KMT5B), ARID1B and CHD8-in multiple cell lines from different donors, using single-cell RNA-sequencing (scRNA-seq) analysis of more than 745,000 cells and proteomic analysis of individual organoids, to identify phenotypic convergence. Each of the three mutations confers asynchronous development of two main cortical neuronal lineages-γ-aminobutyric-acid-releasing (GABAergic) neurons and deep-layer excitatory projection neurons-but acts through largely distinct molecular pathways. Although these phenotypes are consistent across cell lines, their expressivity is influenced by the individual genomic context, in a manner that is dependent on both the risk gene and the developmental defect. Calcium imaging in intact organoids shows that these early-stage developmental changes are followed by abnormal circuit activity. This research uncovers cell-type-specific neurodevelopmental abnormalities that are shared across ASD risk genes and are finely modulated by human genomic context, finding convergence in the neurobiological basis of how different risk genes contribute to ASD pathology.


Asunto(s)
Trastorno del Espectro Autista , Predisposición Genética a la Enfermedad , Neuronas , Trastorno del Espectro Autista/genética , Trastorno del Espectro Autista/metabolismo , Trastorno del Espectro Autista/patología , Corteza Cerebral/citología , Proteínas de Unión al ADN/genética , Neuronas GABAérgicas/metabolismo , Neuronas GABAérgicas/patología , N-Metiltransferasa de Histona-Lisina/genética , Humanos , Neuronas/clasificación , Neuronas/metabolismo , Neuronas/patología , Organoides/citología , Proteómica , RNA-Seq , Análisis de la Célula Individual , Factores de Transcripción/genética
10.
Development ; 151(10)2024 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-38775708

RESUMEN

In utero infection and maternal inflammation can adversely impact fetal brain development. Maternal systemic illness, even in the absence of direct fetal brain infection, is associated with an increased risk of neuropsychiatric disorders in affected offspring. The cell types mediating the fetal brain response to maternal inflammation are largely unknown, hindering the development of novel treatment strategies. Here, we show that microglia, the resident phagocytes of the brain, highly express receptors for relevant pathogens and cytokines throughout embryonic development. Using a rodent maternal immune activation (MIA) model in which polyinosinic:polycytidylic acid is injected into pregnant mice, we demonstrate long-lasting transcriptional changes in fetal microglia that persist into postnatal life. We find that MIA induces widespread gene expression changes in neuronal and non-neuronal cells; importantly, these responses are abolished by selective genetic deletion of microglia, indicating that microglia are required for the transcriptional response of other cortical cell types to MIA. These findings demonstrate that microglia play a crucial durable role in the fetal response to maternal inflammation, and should be explored as potential therapeutic cell targets.


Asunto(s)
Encéfalo , Inflamación , Microglía , Poli I-C , Animales , Microglía/metabolismo , Microglía/inmunología , Femenino , Embarazo , Ratones , Encéfalo/patología , Encéfalo/inmunología , Encéfalo/metabolismo , Inflamación/patología , Inflamación/genética , Poli I-C/farmacología , Feto , Ratones Endogámicos C57BL , Regulación del Desarrollo de la Expresión Génica , Neuronas/metabolismo
11.
Nature ; 583(7818): 819-824, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32699411

RESUMEN

The thalamic reticular nucleus (TRN), the major source of thalamic inhibition, regulates thalamocortical interactions that are critical for sensory processing, attention and cognition1-5. TRN dysfunction has been linked to sensory abnormality, attention deficit and sleep disturbance across multiple neurodevelopmental disorders6-9. However, little is known about the organizational principles that underlie its divergent functions. Here we performed an integrative study linking single-cell molecular and electrophysiological features of the mouse TRN to connectivity and systems-level function. We found that cellular heterogeneity in the TRN is characterized by a transcriptomic gradient of two negatively correlated gene-expression profiles, each containing hundreds of genes. Neurons in the extremes of this transcriptomic gradient express mutually exclusive markers, exhibit core or shell-like anatomical structure and have distinct electrophysiological properties. The two TRN subpopulations make differential connections with the functionally distinct first-order and higher-order thalamic nuclei to form molecularly defined TRN-thalamus subnetworks. Selective perturbation of the two subnetworks in vivo revealed their differential role in regulating sleep. In sum, our study provides a comprehensive atlas of TRN neurons at single-cell resolution and links molecularly defined subnetworks to the functional organization of thalamocortical circuits.


Asunto(s)
Redes Reguladoras de Genes , Núcleos Talámicos/citología , Núcleos Talámicos/metabolismo , Animales , Análisis por Conglomerados , Femenino , Perfilación de la Expresión Génica , Hibridación Fluorescente in Situ , Metaloendopeptidasas/metabolismo , Ratones , Vías Nerviosas , Neuronas/metabolismo , Osteopontina/metabolismo , Técnicas de Placa-Clamp , RNA-Seq , Análisis de la Célula Individual , Sueño/genética , Sueño/fisiología , Núcleos Talámicos/fisiología , Transcriptoma
12.
Hum Mol Genet ; 32(18): 2773-2786, 2023 09 05.
Artículo en Inglés | MEDLINE | ID: mdl-37384417

RESUMEN

De novo heterozygous loss-of-function mutations in phosphatase and tensin homolog (PTEN) are strongly associated with autism spectrum disorders; however, it is unclear how heterozygous mutations in this gene affect different cell types during human brain development and how these effects vary across individuals. Here, we used human cortical organoids from different donors to identify cell-type specific developmental events that are affected by heterozygous mutations in PTEN. We profiled individual organoids by single-cell RNA-seq, proteomics and spatial transcriptomics and revealed abnormalities in developmental timing in human outer radial glia progenitors and deep-layer cortical projection neurons, which varied with the donor genetic background. Calcium imaging in intact organoids showed that both accelerated and delayed neuronal development phenotypes resulted in similar abnormal activity of local circuits, irrespective of genetic background. The work reveals donor-dependent, cell-type specific developmental phenotypes of PTEN heterozygosity that later converge on disrupted neuronal activity.


Asunto(s)
Trastorno del Espectro Autista , Neuronas , Humanos , Neuronas/metabolismo , Diferenciación Celular , Organoides/metabolismo , Trastorno del Espectro Autista/genética , Mutación , Fosfohidrolasa PTEN/genética
13.
Nature ; 570(7762): 523-527, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-31168097

RESUMEN

Experimental models of the human brain are needed for basic understanding of its development and disease1. Human brain organoids hold unprecedented promise for this purpose; however, they are plagued by high organoid-to-organoid variability2,3. This has raised doubts as to whether developmental processes of the human brain can occur outside the context of embryogenesis with a degree of reproducibility that is comparable to the endogenous tissue. Here we show that an organoid model of the dorsal forebrain can reliably generate a rich diversity of cell types appropriate for the human cerebral cortex. We performed single-cell RNA-sequencing analysis of 166,242 cells isolated from 21 individual organoids, finding that 95% of the organoids generate a virtually indistinguishable compendium of cell types, following similar developmental trajectories and with a degree of organoid-to-organoid variability comparable to that of individual endogenous brains. Furthermore, organoids derived from different stem cell lines show consistent reproducibility in the cell types produced. The data demonstrate that reproducible development of the complex cellular diversity of the central nervous system does not require the context of the embryo, and that establishment of terminal cell identity is a highly constrained process that can emerge from diverse stem cell origins and growth environments.


Asunto(s)
Corteza Cerebral/citología , Organoides/citología , Técnicas de Cultivo de Tejidos , Línea Celular , Corteza Cerebral/metabolismo , Femenino , Feto/citología , Feto/metabolismo , Humanos , Células Madre Pluripotentes Inducidas/citología , Masculino , Organoides/metabolismo , Prosencéfalo/citología , Prosencéfalo/metabolismo , RNA-Seq , Reproducibilidad de los Resultados , Análisis de la Célula Individual , Factores de Tiempo , Técnicas de Cultivo de Tejidos/normas , Transcriptoma/genética
14.
Proc Natl Acad Sci U S A ; 118(13)2021 03 30.
Artículo en Inglés | MEDLINE | ID: mdl-33766915

RESUMEN

Microglial-derived inflammation has been linked to a broad range of neurodegenerative and neuropsychiatric conditions, including amyotrophic lateral sclerosis (ALS). Using single-cell RNA sequencing, a class of Disease-Associated Microglia (DAMs) have been characterized in neurodegeneration. However, the DAM phenotype alone is insufficient to explain the functional complexity of microglia, particularly with regard to regulating inflammation that is a hallmark of many neurodegenerative diseases. Here, we identify a subclass of microglia in mouse models of ALS which we term RIPK1-Regulated Inflammatory Microglia (RRIMs). RRIMs show significant up-regulation of classical proinflammatory pathways, including increased levels of Tnf and Il1b RNA and protein. We find that RRIMs are highly regulated by TNFα signaling and that the prevalence of these microglia can be suppressed by inhibiting receptor-interacting protein kinase 1 (RIPK1) activity downstream of the TNF receptor 1. These findings help to elucidate a mechanism by which RIPK1 kinase inhibition has been shown to provide therapeutic benefit in mouse models of ALS and may provide an additional biomarker for analysis in ongoing phase 2 clinical trials of RIPK1 inhibitors in ALS.


Asunto(s)
Esclerosis Amiotrófica Lateral/enzimología , Inflamación/enzimología , Microglía/enzimología , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Esclerosis Amiotrófica Lateral/genética , Esclerosis Amiotrófica Lateral/patología , Animales , Proteínas de Ciclo Celular/genética , Modelos Animales de Enfermedad , Interleucina-1beta/metabolismo , Proteínas de Transporte de Membrana/genética , Ratones , Ratones Mutantes , Microglía/patología , Proteína Serina-Treonina Quinasas de Interacción con Receptores/antagonistas & inhibidores , Proteína Serina-Treonina Quinasas de Interacción con Receptores/genética , Análisis de la Célula Individual , Superóxido Dismutasa-1/genética , Transcriptoma , Factor de Necrosis Tumoral alfa/metabolismo
15.
Nat Methods ; 15(12): 1126, 2018 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-30459407

RESUMEN

The original version of this paper contained an incorrect primer sequence. In the Methods subsection "Rampage libraries," the text for modification 3 stated that the reverse primer used for library indexing was 5'-CAAGCAGAAGACGGCATACGAGATXXXXXXXXGTGACTGGAGT-3'. The correct sequence of the oligonucleotide used is 5'-CAAGCAGAAGACGGCATACGAGATXXXXXXXXGTGACTGGAGTTCAGACGTGTGCTCTTCCGATCT-3'. This error has been corrected in the PDF and HTML versions of the paper.

16.
Nat Methods ; 15(7): 505-511, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29867192

RESUMEN

Specialized RNA-seq methods are required to identify the 5' ends of transcripts, which are critical for studies of gene regulation, but these methods have not been systematically benchmarked. We directly compared six such methods, including the performance of five methods on a single human cellular RNA sample and a new spike-in RNA assay that helps circumvent challenges resulting from uncertainties in annotation and RNA processing. We found that the 'cap analysis of gene expression' (CAGE) method performed best for mRNA and that most of its unannotated peaks were supported by evidence from other genomic methods. We applied CAGE to eight brain-related samples and determined sample-specific transcription start site (TSS) usage, as well as a transcriptome-wide shift in TSS usage between fetal and adult brain.


Asunto(s)
ARN/química , Análisis de Secuencia de ARN/métodos , Secuencia de Bases , Encéfalo , Células Madre Embrionarias , Biblioteca de Genes , Humanos , ARN/genética , ARN/metabolismo
17.
Proc Natl Acad Sci U S A ; 114(41): E8788-E8797, 2017 10 10.
Artículo en Inglés | MEDLINE | ID: mdl-28904096

RESUMEN

Dysfunction of microglia is known to play an important role in Alzheimer's disease (AD). Here, we investigated the role of RIPK1 in microglia mediating the pathogenesis of AD. RIPK1 is highly expressed by microglial cells in human AD brains. Using the amyloid precursor protein (APP)/presenilin 1 (PS1) transgenic mouse model, we found that inhibition of RIPK1, using both pharmacological and genetic means, reduced amyloid burden, the levels of inflammatory cytokines, and memory deficits. Furthermore, inhibition of RIPK1 promoted microglial degradation of Aß in vitro. We characterized the transcriptional profiles of adult microglia from APP/PS1 mice and identified a role for RIPK1 in regulating the microglial expression of CH25H and Cst7, a marker for disease-associated microglia (DAM), which encodes an endosomal/lysosomal cathepsin inhibitor named Cystatin F. We present evidence that RIPK1-mediated induction of Cst7 leads to an impairment in the lysosomal pathway. These data suggest that RIPK1 may mediate a critical checkpoint in the transition to the DAM state. Together, our study highlights a non-cell death mechanism by which the activation of RIPK1 mediates the induction of a DAM phenotype, including an inflammatory response and a reduction in phagocytic activity, and connects RIPK1-mediated transcription in microglia to the etiology of AD. Our results support that RIPK1 is an important therapeutic target for the treatment of AD.


Asunto(s)
Enfermedad de Alzheimer/patología , Biomarcadores/metabolismo , Microglía/patología , Presenilina-1/fisiología , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/metabolismo , Animales , Células Cultivadas , Citocinas/metabolismo , Modelos Animales de Enfermedad , Perfilación de la Expresión Génica , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Microglía/metabolismo , Fenotipo , Proteína Serina-Treonina Quinasas de Interacción con Receptores/genética
18.
Nature ; 498(7453): 236-40, 2013 Jun 13.
Artículo en Inglés | MEDLINE | ID: mdl-23685454

RESUMEN

Recent molecular studies have shown that, even when derived from a seemingly homogenous population, individual cells can exhibit substantial differences in gene expression, protein levels and phenotypic output, with important functional consequences. Existing studies of cellular heterogeneity, however, have typically measured only a few pre-selected RNAs or proteins simultaneously, because genomic profiling methods could not be applied to single cells until very recently. Here we use single-cell RNA sequencing to investigate heterogeneity in the response of mouse bone-marrow-derived dendritic cells (BMDCs) to lipopolysaccharide. We find extensive, and previously unobserved, bimodal variation in messenger RNA abundance and splicing patterns, which we validate by RNA-fluorescence in situ hybridization for select transcripts. In particular, hundreds of key immune genes are bimodally expressed across cells, surprisingly even for genes that are very highly expressed at the population average. Moreover, splicing patterns demonstrate previously unobserved levels of heterogeneity between cells. Some of the observed bimodality can be attributed to closely related, yet distinct, known maturity states of BMDCs; other portions reflect differences in the usage of key regulatory circuits. For example, we identify a module of 137 highly variable, yet co-regulated, antiviral response genes. Using cells from knockout mice, we show that variability in this module may be propagated through an interferon feedback circuit, involving the transcriptional regulators Stat2 and Irf7. Our study demonstrates the power and promise of single-cell genomics in uncovering functional diversity between cells and in deciphering cell states and circuits.


Asunto(s)
Células Dendríticas/metabolismo , Perfilación de la Expresión Génica , Regulación de la Expresión Génica/inmunología , Empalme del ARN/inmunología , Análisis de la Célula Individual , Transcriptoma/genética , Animales , Células de la Médula Ósea/citología , Células de la Médula Ósea/inmunología , Células Dendríticas/citología , Células Dendríticas/inmunología , Hibridación Fluorescente in Situ , Factor 7 Regulador del Interferón , Interferones/inmunología , Lipopolisacáridos/inmunología , Ratones , Ratones Noqueados , Isoformas de Proteínas/genética , ARN Mensajero/análisis , ARN Mensajero/genética , Reproducibilidad de los Resultados , Factor de Transcripción STAT2 , Análisis de Secuencia de ARN , Virus/inmunología
19.
Nature ; 496(7445): 311-6, 2013 Apr 18.
Artículo en Inglés | MEDLINE | ID: mdl-23598338

RESUMEN

The discovery of a living coelacanth specimen in 1938 was remarkable, as this lineage of lobe-finned fish was thought to have become extinct 70 million years ago. The modern coelacanth looks remarkably similar to many of its ancient relatives, and its evolutionary proximity to our own fish ancestors provides a glimpse of the fish that first walked on land. Here we report the genome sequence of the African coelacanth, Latimeria chalumnae. Through a phylogenomic analysis, we conclude that the lungfish, and not the coelacanth, is the closest living relative of tetrapods. Coelacanth protein-coding genes are significantly more slowly evolving than those of tetrapods, unlike other genomic features. Analyses of changes in genes and regulatory elements during the vertebrate adaptation to land highlight genes involved in immunity, nitrogen excretion and the development of fins, tail, ear, eye, brain and olfaction. Functional assays of enhancers involved in the fin-to-limb transition and in the emergence of extra-embryonic tissues show the importance of the coelacanth genome as a blueprint for understanding tetrapod evolution.


Asunto(s)
Evolución Biológica , Peces/clasificación , Peces/genética , Genoma/genética , Animales , Animales Modificados Genéticamente , Embrión de Pollo , Secuencia Conservada/genética , Elementos de Facilitación Genéticos/genética , Evolución Molecular , Extremidades/anatomía & histología , Extremidades/crecimiento & desarrollo , Peces/anatomía & histología , Peces/fisiología , Genes Homeobox/genética , Genómica , Inmunoglobulina M/genética , Ratones , Anotación de Secuencia Molecular , Datos de Secuencia Molecular , Filogenia , Alineación de Secuencia , Análisis de Secuencia de ADN , Vertebrados/anatomía & histología , Vertebrados/genética , Vertebrados/fisiología
20.
RNA ; 22(5): 660-6, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-26968626

RESUMEN

The MS2 system has been extensively used to visualize single mRNA molecules in live cells and follow their localization and behavior. In their Letter to the Editor recently published, Garcia and Parker suggest that use of the MS2 system may yield erroneous mRNA localization results due to the accumulation of 3' decay products. Here we cite published works and provide new data which demonstrate that this is not a phenomenon general to endogenously expressed MS2-tagged transcripts, and that some of the results obtained in their study could have arisen from artifacts of gene expression.


Asunto(s)
Aptámeros de Nucleótidos/metabolismo , Proteínas Fúngicas/metabolismo , Unión Proteica , ARN Mensajero/metabolismo
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda