Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
bioRxiv ; 2024 Mar 25.
Artículo en Inglés | MEDLINE | ID: mdl-38585858

RESUMEN

Torpor and hibernation are extreme physiological adaptations of homeotherms associated with pro-longevity effects. Yet the underlying mechanisms of how torpor affects aging, and whether hypothermic and hypometabolic states can be induced to slow aging and increase health span, remain unknown. We demonstrate that the activity of a spatially defined neuronal population in the avMLPA, which has previously been identified as a torpor-regulating brain region, is sufficient to induce a torpor like state (TLS) in mice. Prolonged induction of TLS slows epigenetic aging across multiple tissues and improves health span. We isolate the effects of decreased metabolic rate, long-term caloric restriction, and decreased core body temperature (Tb) on blood epigenetic aging and find that the pro-longevity effect of torpor-like states is mediated by decreased Tb. Taken together, our findings provide novel mechanistic insight into the pro-longevity effects of torpor and hibernation and support the growing body of evidence that Tb is an important mediator of aging processes.

2.
Nature ; 620(7973): 366-373, 2023 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-37468637

RESUMEN

Neurons in the posterior parietal cortex contribute to the execution of goal-directed navigation1 and other decision-making tasks2-4. Although molecular studies have catalogued more than 50 cortical cell types5, it remains unclear what distinct functions they have in this area. Here we identified a molecularly defined subset of somatostatin (Sst) inhibitory neurons that, in the mouse posterior parietal cortex, carry a cell-type-specific error-correction signal for navigation. We obtained repeatable experimental access to these cells using an adeno-associated virus in which gene expression is driven by an enhancer that functions specifically in a subset of Sst cells6. We found that during goal-directed navigation in a virtual environment, this subset of Sst neurons activates in a synchronous pattern that is distinct from the activity of surrounding neurons, including other Sst neurons. Using in vivo two-photon photostimulation and ex vivo paired patch-clamp recordings, we show that nearby cells of this Sst subtype excite each other through gap junctions, revealing a self-excitation circuit motif that contributes to the synchronous activity of this cell type. These cells selectively activate as mice execute course corrections for deviations in their virtual heading during navigation towards a reward location, for both self-induced and experimentally induced deviations. We propose that this subtype of Sst neurons provides a self-reinforcing and cell-type-specific error-correction signal in the posterior parietal cortex that may help with the execution and learning of accurate goal-directed navigation trajectories.


Asunto(s)
Neuronas , Lóbulo Parietal , Animales , Ratones , Aprendizaje , Neuronas/metabolismo , Lóbulo Parietal/citología , Lóbulo Parietal/metabolismo , Objetivos , Somatostatina/metabolismo , Inhibición Neural , Navegación Espacial , Técnicas de Placa-Clamp , Uniones Comunicantes/metabolismo
3.
Elife ; 102021 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-34851821

RESUMEN

The septum is a ventral forebrain structure known to regulate innate behaviors. During embryonic development, septal neurons are produced in multiple proliferative areas from neural progenitors following transcriptional programs that are still largely unknown. Here, we use a combination of single-cell RNA sequencing, histology, and genetic models to address how septal neuron diversity is established during neurogenesis. We find that the transcriptional profiles of septal progenitors change along neurogenesis, coinciding with the generation of distinct neuron types. We characterize the septal eminence, an anatomically distinct and transient proliferative zone composed of progenitors with distinctive molecular profiles, proliferative capacity, and fate potential compared to the rostral septal progenitor zone. We show that Nkx2.1-expressing septal eminence progenitors give rise to neurons belonging to at least three morphological classes, born in temporal cohorts that are distributed across different septal nuclei in a sequential fountain-like pattern. Our study provides insight into the molecular programs that control the sequential production of different neuronal types in the septum, a structure with important roles in regulating mood and motivation.


Asunto(s)
Neurogénesis/genética , Neuronas/fisiología , Tabique del Cerebro/fisiología , Factor Nuclear Tiroideo 1/genética , Transcripción Genética , Animales , Femenino , Perfilación de la Expresión Génica , Masculino , Ratones , Factor Nuclear Tiroideo 1/metabolismo
4.
Nat Neurosci ; 24(3): 437-448, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33542524

RESUMEN

Neuronal activity-dependent gene expression is essential for brain development. Although transcriptional and epigenetic effects of neuronal activity have been explored in mice, such an investigation is lacking in humans. Because alterations in GABAergic neuronal circuits are implicated in neurological disorders, we conducted a comprehensive activity-dependent transcriptional and epigenetic profiling of human induced pluripotent stem cell-derived GABAergic neurons similar to those of the early developing striatum. We identified genes whose expression is inducible after membrane depolarization, some of which have specifically evolved in primates and/or are associated with neurological diseases, including schizophrenia and autism spectrum disorder (ASD). We define the genome-wide profile of human neuronal activity-dependent enhancers, promoters and the transcription factors CREB and CRTC1. We found significant heritability enrichment for ASD in the inducible promoters. Our results suggest that sequence variation within activity-inducible promoters of developing human forebrain GABAergic neurons contributes to ASD risk.


Asunto(s)
Encéfalo/metabolismo , Epigénesis Genética , Neuronas GABAérgicas/metabolismo , Regulación de la Expresión Génica , Estudio de Asociación del Genoma Completo , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Regiones Promotoras Genéticas
5.
Nature ; 583(7814): 115-121, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32528180

RESUMEN

The advent of endothermy, which is achieved through the continuous homeostatic regulation of body temperature and metabolism1,2, is a defining feature of mammalian and avian evolution. However, when challenged by food deprivation or harsh environmental conditions, many mammalian species initiate adaptive energy-conserving survival strategies-including torpor and hibernation-during which their body temperature decreases far below its homeostatic set-point3-5. How homeothermic mammals initiate and regulate these hypothermic states remains largely unknown. Here we show that entry into mouse torpor, a fasting-induced state with a greatly decreased metabolic rate and a body temperature as low as 20 °C6, is regulated by neurons in the medial and lateral preoptic area of the hypothalamus. We show that restimulation of neurons that were activated during a previous bout of torpor is sufficient to initiate the key features of torpor, even in mice that are not calorically restricted. Among these neurons we identify a population of glutamatergic Adcyap1-positive cells, the activity of which accurately determines when mice naturally initiate and exit torpor, and the inhibition of which disrupts the natural process of torpor entry, maintenance and arousal. Taken together, our results reveal a specific neuronal population in the mouse hypothalamus that serves as a core regulator of torpor. This work forms a basis for the future exploration of mechanisms and circuitry that regulate extreme hypothermic and hypometabolic states, and enables genetic access to monitor, initiate, manipulate and study these ancient adaptations of homeotherm biology.


Asunto(s)
Metabolismo Energético/fisiología , Hipotálamo/citología , Vías Nerviosas/fisiología , Neuronas/fisiología , Letargo/fisiología , Animales , Ayuno , Femenino , Privación de Alimentos , Glutamina/metabolismo , Hipotálamo/fisiología , Masculino , Ratones , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/metabolismo
6.
Neuron ; 107(5): 874-890.e8, 2020 09 09.
Artículo en Inglés | MEDLINE | ID: mdl-32589877

RESUMEN

The maturation of the mammalian brain occurs after birth, and this stage of neuronal development is frequently impaired in neurological disorders, such as autism and schizophrenia. However, the mechanisms that regulate postnatal brain maturation are poorly defined. By purifying neuronal subpopulations across brain development in mice, we identify a postnatal switch in the transcriptional regulatory circuits that operates in the maturing mammalian brain. We show that this developmental transition includes the formation of hundreds of cell-type-specific neuronal enhancers that appear to be modulated by neuronal activity. Once selected, these enhancers are active throughout adulthood, suggesting that their formation in early life shapes neuronal identity and regulates mature brain function.


Asunto(s)
Encéfalo/crecimiento & desarrollo , Regulación de la Expresión Génica/fisiología , Neurogénesis/fisiología , Neuronas/fisiología , Animales , Metilación de ADN/fisiología , Ratones , Transcripción Genética/fisiología
7.
Elife ; 82019 09 23.
Artículo en Inglés | MEDLINE | ID: mdl-31545165

RESUMEN

Enhancers are the primary DNA regulatory elements that confer cell type specificity of gene expression. Recent studies characterizing individual enhancers have revealed their potential to direct heterologous gene expression in a highly cell-type-specific manner. However, it has not yet been possible to systematically identify and test the function of enhancers for each of the many cell types in an organism. We have developed PESCA, a scalable and generalizable method that leverages ATAC- and single-cell RNA-sequencing protocols, to characterize cell-type-specific enhancers that should enable genetic access and perturbation of gene function across mammalian cell types. Focusing on the highly heterogeneous mammalian cerebral cortex, we apply PESCA to find enhancers and generate viral reagents capable of accessing and manipulating a subset of somatostatin-expressing cortical interneurons with high specificity. This study demonstrates the utility of this platform for developing new cell-type-specific viral reagents, with significant implications for both basic and translational research.


Asunto(s)
Regulación de la Expresión Génica/efectos de los fármacos , Biología Molecular/métodos , Neuronas/efectos de los fármacos , Neurofisiología/métodos , Proteínas Recombinantes/biosíntesis , Somatostatina/metabolismo , Virus/genética , Animales , Animales Modificados Genéticamente , Corteza Cerebral/fisiología , Genes Reguladores , Vectores Genéticos , Interneuronas/fisiología , Ratones , Proteínas Recombinantes/genética
8.
Neuron ; 102(2): 390-406.e9, 2019 04 17.
Artículo en Inglés | MEDLINE | ID: mdl-30846309

RESUMEN

Neuronal activity-dependent transcription is tuned to ensure precise gene induction during periods of heightened synaptic activity, allowing for appropriate responses of activated neurons within neural circuits. The consequences of aberrant induction of activity-dependent genes on neuronal physiology are not yet clear. Here, we demonstrate that, in the absence of synaptic excitation, the basic-helix-loop-helix (bHLH)-PAS family transcription factor ARNT2 recruits the NCoR2 co-repressor complex to suppress neuronal activity-dependent regulatory elements and maintain low basal levels of inducible genes. This restricts inhibition of excitatory neurons, maintaining them in a state that is receptive to future sensory stimuli. By contrast, in response to heightened neuronal activity, ARNT2 recruits the neuronal-specific bHLH-PAS factor NPAS4 to activity-dependent regulatory elements to induce transcription and thereby increase somatic inhibitory input. Thus, the interplay of bHLH-PAS complexes at activity-dependent regulatory elements maintains temporal control of activity-dependent gene expression and scales somatic inhibition with circuit activity.


Asunto(s)
Translocador Nuclear del Receptor de Aril Hidrocarburo/metabolismo , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Regulación de la Expresión Génica , Neuronas/metabolismo , Co-Represor 2 de Receptor Nuclear/metabolismo , Animales , Ratones , Inhibición Neural , Elementos Reguladores de la Transcripción , Activación Transcripcional
9.
Nat Neurosci ; 21(12): 1670-1679, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30455458

RESUMEN

In females with X-linked genetic disorders, wild-type and mutant cells coexist within brain tissue because of X-chromosome inactivation, posing challenges for interpreting the effects of X-linked mutant alleles on gene expression. We present a single-nucleus RNA sequencing approach that resolves mosaicism by using single-nucleotide polymorphisms in genes expressed in cis with the X-linked mutation to determine which nuclei express the mutant allele even when the mutant gene is not detected. This approach enables gene expression comparisons between mutant and wild-type cells within the same individual, eliminating variability introduced by comparisons to controls with different genetic backgrounds. We apply this approach to mosaic female mouse models and humans with Rett syndrome, an X-linked neurodevelopmental disorder caused by mutations in the gene encoding the methyl-DNA-binding protein MECP2, and observe that cell-type-specific DNA methylation predicts the degree of gene upregulation in MECP2-mutant neurons. This approach can be broadly applied to study gene expression in mosaic X-linked disorders.


Asunto(s)
Encéfalo/metabolismo , Proteína 2 de Unión a Metil-CpG/genética , Síndrome de Rett/genética , Alelos , Metilación de ADN , Femenino , Humanos , Proteína 2 de Unión a Metil-CpG/metabolismo , Mosaicismo , Mutación , Neuronas/metabolismo , Polimorfismo de Nucleótido Simple , Síndrome de Rett/metabolismo , Análisis de Secuencia de ARN
10.
Neuron ; 99(3): 525-539.e10, 2018 08 08.
Artículo en Inglés | MEDLINE | ID: mdl-30033152

RESUMEN

Sensory experience influences the establishment of neural connectivity through molecular mechanisms that remain unclear. Here, we employ single-nucleus RNA sequencing to investigate the contribution of sensory-driven gene expression to synaptic refinement in the dorsal lateral geniculate nucleus of the thalamus, a region of the brain that processes visual information. We find that visual experience induces the expression of the cytokine receptor Fn14 in excitatory thalamocortical neurons. By combining electrophysiological and structural techniques, we show that Fn14 is dispensable for early phases of refinement mediated by spontaneous activity but that Fn14 is essential for refinement during a later, experience-dependent period of development. Refinement deficits in mice lacking Fn14 are associated with functionally weaker and structurally smaller retinogeniculate inputs, indicating that Fn14 mediates both functional and anatomical rearrangements in response to sensory experience. These findings identify Fn14 as a molecular link between sensory-driven gene expression and vision-sensitive refinement in the brain.


Asunto(s)
Cuerpos Geniculados/metabolismo , Células Ganglionares de la Retina/metabolismo , Receptor de TWEAK/biosíntesis , Percepción Visual/fisiología , Animales , Femenino , Expresión Génica , Cuerpos Geniculados/crecimiento & desarrollo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Tracto Óptico/crecimiento & desarrollo , Tracto Óptico/metabolismo , Retina/metabolismo , Receptor de TWEAK/genética
11.
Nat Neurosci ; 21(7): 1017, 2018 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-29752482

RESUMEN

In the version of this article initially published, the x-axis labels in Fig. 3c read Vglut, Gad1/2, Aldh1l1 and Pecam1; they should have read Vglut+, Gad1/2+, Aldh1l1+ and Pecam1+. In Fig. 4, the range values were missing from the color scales; they are, from left to right, 4-15, 0-15, 4-15 and 0-15 in Fig. 4a and 4-15, 4-15 and 4-8 in Fig. 4h. In the third paragraph of the main text, the phrase reading "Previous approaches have analyzed a limited number of inhibitory cell types, thus masking the full diversity of excitatory populations" should have read "Previous approaches have analyzed a limited number of inhibitory cell types and masked the full diversity of excitatory populations." In the second paragraph of Results section "Diversity of experience-regulated ERGs," the phrase reading "thus suggesting considerable divergence within the gene expression program responding to early stimuli" should have read "thus suggesting considerable divergence within the early stimulus-responsive gene expression program." In the fourth paragraph of Results section "Excitatory neuronal LRGs," the sentence reading "The anatomical organization of these cell types into sublayers, coupled with divergent transcriptional responses to a sensory stimulus, suggested previously unappreciated functional subdivisions located within the laminae of the mouse visual cortex and resembling the cytoarchitecture in higher mammals" should have read "The anatomical organization of these cell types into sublayers, coupled with divergent transcriptional responses to a sensory stimulus, suggests previously unappreciated functional subdivisions located within the laminae of the mouse visual cortex, resembling the cytoarchitecture in higher mammals." In the last sentence of the Results, "sensory-responsive genes" should have read "sensory-stimulus-responsive genes." The errors have been corrected in the HTML and PDF versions of the article.

12.
Proc Natl Acad Sci U S A ; 115(5): E1051-E1060, 2018 01 30.
Artículo en Inglés | MEDLINE | ID: mdl-29343640

RESUMEN

Coordinated changes in gene expression underlie the early patterning and cell-type specification of the central nervous system. However, much less is known about how such changes contribute to later stages of circuit assembly and refinement. In this study, we employ single-cell RNA sequencing to develop a detailed, whole-transcriptome resource of gene expression across four time points in the developing dorsal lateral geniculate nucleus (LGN), a visual structure in the brain that undergoes a well-characterized program of postnatal circuit development. This approach identifies markers defining the major LGN cell types, including excitatory relay neurons, oligodendrocytes, astrocytes, microglia, and endothelial cells. Most cell types exhibit significant transcriptional changes across development, dynamically expressing genes involved in distinct processes including retinotopic mapping, synaptogenesis, myelination, and synaptic refinement. Our data suggest that genes associated with synapse and circuit development are expressed in a larger proportion of nonneuronal cell types than previously appreciated. Furthermore, we used this single-cell expression atlas to identify the Prkcd-Cre mouse line as a tool for selective manipulation of relay neurons during a late stage of sensory-driven synaptic refinement. This transcriptomic resource provides a cellular map of gene expression across several cell types of the LGN, and offers insight into the molecular mechanisms of circuit development in the postnatal brain.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Cuerpos Geniculados/embriología , Cuerpos Geniculados/fisiología , Neuronas/fisiología , Sinapsis/fisiología , Transcriptoma , Animales , Axones/fisiología , Encéfalo/embriología , Perfilación de la Expresión Génica , Ratones , Microscopía Electrónica de Rastreo , Neurogénesis , Retina/fisiología , Análisis de Secuencia de ARN , Programas Informáticos , Vías Visuales/fisiología
13.
Nat Neurosci ; 21(1): 120-129, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29230054

RESUMEN

Activity-dependent transcriptional responses shape cortical function. However, a comprehensive understanding of the diversity of these responses across the full range of cortical cell types, and how these changes contribute to neuronal plasticity and disease, is lacking. To investigate the breadth of transcriptional changes that occur across cell types in the mouse visual cortex after exposure to light, we applied high-throughput single-cell RNA sequencing. We identified significant and divergent transcriptional responses to stimulation in each of the 30 cell types characterized, thus revealing 611 stimulus-responsive genes. Excitatory pyramidal neurons exhibited inter- and intralaminar heterogeneity in the induction of stimulus-responsive genes. Non-neuronal cells showed clear transcriptional responses that may regulate experience-dependent changes in neurovascular coupling and myelination. Together, these results reveal the dynamic landscape of the stimulus-dependent transcriptional changes occurring across cell types in the visual cortex; these changes are probably critical for cortical function and may be sites of deregulation in developmental brain disorders.


Asunto(s)
Neuroglía/fisiología , Neuronas/fisiología , Transcripción Genética/fisiología , Transcriptoma/fisiología , Corteza Visual/citología , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Regulación de la Expresión Génica/fisiología , Ontología de Genes , Luz , Masculino , Ratones , Ratones Endogámicos C57BL , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Inhibición Neural/fisiología , Neuronas/citología , Acoplamiento Neurovascular/fisiología , Estimulación Luminosa , Proteínas Proto-Oncogénicas c-fos/metabolismo , Transducción de Señal/fisiología , Análisis de la Célula Individual/métodos , Estadísticas no Paramétricas , Vías Visuales
14.
Cell ; 171(5): 1151-1164.e16, 2017 Nov 16.
Artículo en Inglés | MEDLINE | ID: mdl-29056337

RESUMEN

In mammals, the environment plays a critical role in promoting the final steps in neuronal development during the early postnatal period. While epigenetic factors are thought to contribute to this process, the underlying molecular mechanisms remain poorly understood. Here, we show that in the brain during early life, the DNA methyltransferase DNMT3A transiently binds across transcribed regions of lowly expressed genes, and its binding specifies the pattern of DNA methylation at CA sequences (mCA) within these genes. We find that DNMT3A occupancy and mCA deposition within the transcribed regions of genes is negatively regulated by gene transcription and may be modified by early-life experience. Once deposited, mCA is bound by the methyl-DNA-binding protein MECP2 and functions in a rheostat-like manner to fine-tune the cell-type-specific transcription of genes that are critical for brain function.


Asunto(s)
ADN (Citosina-5-)-Metiltransferasas/genética , ADN (Citosina-5-)-Metiltransferasas/metabolismo , Metilación de ADN , Epigénesis Genética , Neuronas/metabolismo , Animales , Encéfalo/citología , Encéfalo/metabolismo , ADN Metiltransferasa 3A , Regulación del Desarrollo de la Expresión Génica , Humanos , Proteína 2 de Unión a Metil-CpG , Ratones , Transcripción Genética , Activación Transcripcional
15.
Cell ; 165(7): 1570-1571, 2016 06 16.
Artículo en Inglés | MEDLINE | ID: mdl-27315474

RESUMEN

Understanding how genes within cells, and cells within circuits, function together to produce the extraordinary repertoire of animal behaviors is arguably one of the most challenging undertakings in neuroscience. Two papers in this issue move toward this goal via 3D imaging of active neurons across the entire mouse brain.


Asunto(s)
Encéfalo , Neuronas , Animales , Conducta Animal , Neurociencias
16.
Neuron ; 85(2): 275-288, 2015 Jan 21.
Artículo en Inglés | MEDLINE | ID: mdl-25556833

RESUMEN

Neuronal development requires a complex choreography of transcriptional decisions to obtain specific cellular identities. Realizing the ultimate goal of identifying genome-wide signatures that define and drive specific neuronal fates has been hampered by enormous complexity in both time and space during development. Here, we have paired high-throughput purification of pyramidal neuron subclasses with deep profiling of spatiotemporal transcriptional dynamics during corticogenesis to resolve lineage choice decisions. We identified numerous features ranging from spatial and temporal usage of alternative mRNA isoforms and promoters to a host of mRNA genes modulated during fate specification. Notably, we uncovered numerous long noncoding RNAs with restricted temporal and cell-type-specific expression. To facilitate future exploration, we provide an interactive online database to enable multidimensional data mining and dissemination. This multifaceted study generates a powerful resource and informs understanding of the transcriptional regulation underlying pyramidal neuron diversity in the neocortex.


Asunto(s)
Diferenciación Celular/genética , Regulación del Desarrollo de la Expresión Génica/genética , Neocórtex/metabolismo , Células Piramidales/metabolismo , ARN Largo no Codificante/genética , ARN Mensajero/genética , Transcriptoma , Animales , Secuencia de Bases , Cuerpo Calloso/citología , Citometría de Flujo , Perfilación de la Expresión Génica , Proteínas de Unión a la Región de Fijación a la Matriz/metabolismo , Ratones , Datos de Secuencia Molecular , Neuronas Motoras , Neurogénesis/genética , Neuronas/metabolismo , Tractos Piramidales/citología , Proteínas Represoras/metabolismo , Factores de Transcripción/metabolismo , Proteínas Supresoras de Tumor/metabolismo
17.
PLoS One ; 9(3): e89459, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24594682

RESUMEN

Transcriptional profiling is a key technique in the study of cell biology that is limited by the availability of reagents to uniquely identify specific cell types and isolate high quality RNA from them. We report a Method for Analyzing RNA following Intracellular Sorting (MARIS) that generates high quality RNA for transcriptome profiling following cellular fixation, intracellular immunofluorescent staining and FACS. MARIS can therefore be used to isolate high quality RNA from many otherwise inaccessible cell types simply based on immunofluorescent tagging of unique intracellular proteins. As proof of principle, we isolate RNA from sorted human embryonic stem cell-derived insulin-expressing cells as well as adult human ß cells. MARIS is a basic molecular biology technique that could be used across several biological disciplines.


Asunto(s)
ARN/análisis , Células Cultivadas , Células Madre Embrionarias/química , Citometría de Flujo , Humanos , Islotes Pancreáticos/química , Transcripción Genética
18.
Proc Natl Acad Sci U S A ; 111(8): 3038-43, 2014 Feb 25.
Artículo en Inglés | MEDLINE | ID: mdl-24516164

RESUMEN

Human pluripotent stem cells (hPSCs) have the potential to generate any human cell type, and one widely recognized goal is to make pancreatic ß cells. To this end, comparisons between differentiated cell types produced in vitro and their in vivo counterparts are essential to validate hPSC-derived cells. Genome-wide transcriptional analysis of sorted insulin-expressing (INS(+)) cells derived from three independent hPSC lines, human fetal pancreata, and adult human islets points to two major conclusions: (i) Different hPSC lines produce highly similar INS(+) cells and (ii) hPSC-derived INS(+) (hPSC-INS(+)) cells more closely resemble human fetal ß cells than adult ß cells. This study provides a direct comparison of transcriptional programs between pure hPSC-INS(+) cells and true ß cells and provides a catalog of genes whose manipulation may convert hPSC-INS(+) cells into functional ß cells.


Asunto(s)
Diferenciación Celular/fisiología , Células Secretoras de Insulina/citología , Páncreas/citología , Células Madre Pluripotentes/citología , Adulto , Diferenciación Celular/genética , Feto/citología , Feto/metabolismo , Citometría de Flujo , Perfilación de la Expresión Génica , Humanos , Células Secretoras de Insulina/metabolismo , Análisis por Micromatrices , Células Madre Pluripotentes/metabolismo
19.
Nat Biotechnol ; 30(3): 261-4, 2012 02 26.
Artículo en Inglés | MEDLINE | ID: mdl-22371083

RESUMEN

Insulin-expressing cells that have been differentiated from human pluripotent stem cells in vitro lack the glucose responsiveness characteristic of mature beta cells. Beta-cell maturation in mice was studied to find genetic markers that enable screens for factors that induce bona fide beta cells in vitro. We find that functional beta-cell maturation is marked by an increase in the glucose threshold for insulin secretion and by expression of the gene urocortin 3.


Asunto(s)
Glucosa/metabolismo , Células Secretoras de Insulina/citología , Urocortinas/metabolismo , Animales , Diferenciación Celular , Humanos , Insulina/metabolismo , Secreción de Insulina , Células Secretoras de Insulina/metabolismo , Ratones , Ratones Noqueados , Células Madre Pluripotentes/citología , Urocortinas/deficiencia , Urocortinas/genética
20.
Proc Natl Acad Sci U S A ; 104(42): 16498-503, 2007 Oct 16.
Artículo en Inglés | MEDLINE | ID: mdl-17940045

RESUMEN

Kutznerides, actinomycete-derived cyclic depsipetides, consist of six nonproteinogenic residues, including a highly oxygenated tricyclic hexahydropyrroloindole, a chlorinated piperazic acid, 2-(1-methylcyclopropyl)-glycine, a beta-branched-hydroxy acid, and 3-hydroxy glutamic acid, for which biosynthetic logic has not been elucidated. Herein we describe the biosynthetic gene cluster for the kutzneride family, identified by degenerate primer PCR for halogenating enzymes postulated to be involved in biosyntheses of these unusual monomers. The 56-kb gene cluster encodes a series of six nonribosomal peptide synthetase (NRPS) modules distributed over three proteins and a variety of tailoring enzymes, including both mononuclear nonheme iron and two flavin-dependent halogenases, and an array of oxygen transfer catalysts. The sequence and organization of NRPS genes support incorporation of the unusual monomer units into the densely functionalized scaffold of kutznerides. Our work provides insight into the formation of this intriguing class of compounds and provides a foundation for elucidating the timing and mechanisms of their biosynthesis.


Asunto(s)
Actinomycetales/genética , Actinomycetales/metabolismo , Depsipéptidos/biosíntesis , Depsipéptidos/genética , Familia de Multigenes , Adenosina Monofosfato/química , Adenosina Monofosfato/metabolismo , Secuencia de Aminoácidos , Clonación Molecular , Depsipéptidos/química , Datos de Secuencia Molecular
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...