Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Nature ; 614(7949): 732-741, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36792830

RESUMEN

Neuronal activity is crucial for adaptive circuit remodelling but poses an inherent risk to the stability of the genome across the long lifespan of postmitotic neurons1-5. Whether neurons have acquired specialized genome protection mechanisms that enable them to withstand decades of potentially damaging stimuli during periods of heightened activity is unknown. Here we identify an activity-dependent DNA repair mechanism in which a new form of the NuA4-TIP60 chromatin modifier assembles in activated neurons around the inducible, neuronal-specific transcription factor NPAS4. We purify this complex from the brain and demonstrate its functions in eliciting activity-dependent changes to neuronal transcriptomes and circuitry. By characterizing the landscape of activity-induced DNA double-strand breaks in the brain, we show that NPAS4-NuA4 binds to recurrently damaged regulatory elements and recruits additional DNA repair machinery to stimulate their repair. Gene regulatory elements bound by NPAS4-NuA4 are partially protected against age-dependent accumulation of somatic mutations. Impaired NPAS4-NuA4 signalling leads to a cascade of cellular defects, including dysregulated activity-dependent transcriptional responses, loss of control over neuronal inhibition and genome instability, which all culminate to reduce organismal lifespan. In addition, mutations in several components of the NuA4 complex are reported to lead to neurodevelopmental and autism spectrum disorders. Together, these findings identify a neuronal-specific complex that couples neuronal activity directly to genome preservation, the disruption of which may contribute to developmental disorders, neurodegeneration and ageing.


Asunto(s)
Encéfalo , Reparación del ADN , Complejos Multiproteicos , Neuronas , Sinapsis , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico , Encéfalo/metabolismo , Roturas del ADN de Doble Cadena , Regulación de la Expresión Génica , Lisina Acetiltransferasa 5/metabolismo , Complejos Multiproteicos/metabolismo , Neuronas/metabolismo , Sinapsis/metabolismo , Mutación , Longevidad/genética , Genoma , Envejecimiento/genética , Enfermedades Neurodegenerativas
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.
Neuron ; 109(20): 3239-3251.e7, 2021 10 20.
Artículo en Inglés | MEDLINE | ID: mdl-34478631

RESUMEN

Human accelerated regions (HARs) are the fastest-evolving regions of the human genome, and many are hypothesized to function as regulatory elements that drive human-specific gene regulatory programs. We interrogate the in vitro enhancer activity and in vivo epigenetic landscape of more than 3,100 HARs during human neurodevelopment, demonstrating that many HARs appear to act as neurodevelopmental enhancers and that sequence divergence at HARs has largely augmented their neuronal enhancer activity. Furthermore, we demonstrate PPP1R17 to be a putative HAR-regulated gene that has undergone remarkable rewiring of its cell type and developmental expression patterns between non-primates and primates and between non-human primates and humans. Finally, we show that PPP1R17 slows neural progenitor cell cycle progression, paralleling the cell cycle length increase seen predominantly in primate and especially human neurodevelopment. Our findings establish HARs as key components in rewiring human-specific neurodevelopmental gene regulatory programs and provide an integrated resource to study enhancer activity of specific HARs.


Asunto(s)
Encéfalo/embriología , Regulación del Desarrollo de la Expresión Génica/genética , Redes Reguladoras de Genes/genética , Animales , Evolución Biológica , Epigenómica , Evolución Molecular , Hurones , Humanos , Macaca , Ratones , Pan troglodytes
5.
Nature ; 590(7844): 115-121, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33299180

RESUMEN

Behavioural experiences activate the FOS transcription factor in sparse populations of neurons that are critical for encoding and recalling specific events1-3. However, there is limited understanding of the mechanisms by which experience drives circuit reorganization to establish a network of Fos-activated cells. It is also not known whether FOS is required in this process beyond serving as a marker of recent neural activity and, if so, which of its many gene targets underlie circuit reorganization. Here we demonstrate that when mice engage in spatial exploration of novel environments, perisomatic inhibition of Fos-activated hippocampal CA1 pyramidal neurons by parvalbumin-expressing interneurons is enhanced, whereas perisomatic inhibition by cholecystokinin-expressing interneurons is weakened. This bidirectional modulation of inhibition is abolished when the function of the FOS transcription factor complex is disrupted. Single-cell RNA-sequencing, ribosome-associated mRNA profiling and chromatin analyses, combined with electrophysiology, reveal that FOS activates the transcription of Scg2, a gene that encodes multiple distinct neuropeptides, to coordinate these changes in inhibition. As parvalbumin- and cholecystokinin-expressing interneurons mediate distinct features of pyramidal cell activity4-6, the SCG2-dependent reorganization of inhibitory synaptic input might be predicted to affect network function in vivo. Consistent with this prediction, hippocampal gamma rhythms and pyramidal cell coupling to theta phase are significantly altered in the absence of Scg2. These findings reveal an instructive role for FOS and SCG2 in establishing a network of Fos-activated neurons via the rewiring of local inhibition to form a selectively modulated state. The opposing plasticity mechanisms acting on distinct inhibitory pathways may support the consolidation of memories over time.


Asunto(s)
Red Nerviosa/citología , Red Nerviosa/fisiología , Inhibición Neural , Plasticidad Neuronal/fisiología , Proteínas Proto-Oncogénicas c-fos/metabolismo , Animales , Región CA1 Hipocampal/metabolismo , Colecistoquinina/metabolismo , Conducta Exploratoria/fisiología , Femenino , Ritmo Gamma , Interneuronas/metabolismo , Masculino , Consolidación de la Memoria , Ratones , Parvalbúminas/metabolismo , Células Piramidales/metabolismo , Secretogranina II/genética , Secretogranina II/metabolismo , Navegación Espacial/fisiología , Ritmo Teta
6.
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
7.
Elife ; 82019 07 08.
Artículo en Inglés | MEDLINE | ID: mdl-31282856

RESUMEN

Identifying gene expression programs underlying both cell-type identity and cellular activities (e.g. life-cycle processes, responses to environmental cues) is crucial for understanding the organization of cells and tissues. Although single-cell RNA-Seq (scRNA-Seq) can quantify transcripts in individual cells, each cell's expression profile may be a mixture of both types of programs, making them difficult to disentangle. Here, we benchmark and enhance the use of matrix factorization to solve this problem. We show with simulations that a method we call consensus non-negative matrix factorization (cNMF) accurately infers identity and activity programs, including their relative contributions in each cell. To illustrate the insights this approach enables, we apply it to published brain organoid and visual cortex scRNA-Seq datasets; cNMF refines cell types and identifies both expected (e.g. cell cycle and hypoxia) and novel activity programs, including programs that may underlie a neurosecretory phenotype and synaptogenesis.


Asunto(s)
Encéfalo/metabolismo , Perfilación de la Expresión Génica/métodos , RNA-Seq/métodos , Análisis de la Célula Individual/métodos , Corteza Visual/metabolismo , Algoritmos , Animales , Encéfalo/citología , Simulación por Computador , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Humanos , Ratones , Modelos Genéticos , Organoides/citología , Organoides/metabolismo , Reproducibilidad de los Resultados , Corteza Visual/citología
8.
Nat Neurosci ; 22(7): 1075-1088, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31209379

RESUMEN

Microglia rapidly respond to changes in neural activity and inflammation to regulate synaptic connectivity. The extracellular signals, particularly neuron-derived molecules, that drive these microglial functions at synapses remain a key open question. Here we show that whisker lesioning, known to dampen cortical activity, induces microglia-mediated synapse elimination. This synapse elimination is dependent on signaling by CX3CR1, the receptor for microglial fractalkine (also known as CXCL1), but not complement receptor 3. Furthermore, mice deficient in CX3CL1 have profound defects in synapse elimination. Single-cell RNA sequencing revealed that Cx3cl1 is derived from cortical neurons, and ADAM10, a metalloprotease that cleaves CX3CL1 into a secreted form, is upregulated specifically in layer IV neurons and in microglia following whisker lesioning. Finally, inhibition of ADAM10 phenocopies Cx3cr1-/- and Cx3cl1-/- synapse elimination defects. Together, these results identify neuron-to-microglia signaling necessary for cortical synaptic remodeling and reveal that context-dependent immune mechanisms are utilized to remodel synapses in the mammalian brain.


Asunto(s)
Proteína ADAM10/fisiología , Secretasas de la Proteína Precursora del Amiloide/fisiología , Receptor 1 de Quimiocinas CX3C/fisiología , Quimiocina CX3CL1/fisiología , Proteínas de la Membrana/fisiología , Microglía/fisiología , Corteza Sensoriomotora/fisiopatología , Tacto/fisiología , Vibrisas/lesiones , Proteína ADAM10/antagonistas & inhibidores , Proteína ADAM10/genética , Secretasas de la Proteína Precursora del Amiloide/antagonistas & inhibidores , Secretasas de la Proteína Precursora del Amiloide/genética , Animales , Receptor 1 de Quimiocinas CX3C/deficiencia , Receptor 1 de Quimiocinas CX3C/genética , Recuento de Células , Femenino , Regulación de la Expresión Génica , Masculino , Proteínas de la Membrana/antagonistas & inhibidores , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Técnicas Analíticas Microfluídicas , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Corteza Sensoriomotora/metabolismo , Corteza Sensoriomotora/patología , Transducción de Señal/fisiología , Análisis de la Célula Individual , Transcriptoma , Vibrisas/fisiología
9.
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
10.
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
11.
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
12.
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.

13.
J Neurosci ; 38(25): 5750-5758, 2018 06 20.
Artículo en Inglés | MEDLINE | ID: mdl-29802202

RESUMEN

Short-term synaptic plasticity contributes to many computations in the brain and allows synapses to keep a finite record of recent activity. Here we have investigated the mechanisms underlying an intriguing form of short-term plasticity termed labile LTP, at hippocampal and PFC synapses in male rats and male and female mice. In the hippocampus, labile LTP is triggered by high-frequency activation of presynaptic axons and is rapidly discharged with further activation of those axons. However, if the synapses are quiescent, they remain potentiated until further presynaptic activation. To distinguish labile LTP from NMDAR-dependent forms of potentiation, we blocked NMDARs in all experiments. Labile LTP was synapse-specific and was accompanied by a decreased paired pulse ratio, consistent with an increased release probability. Presynaptic Ca2+ and protein kinase activation during the tetanus appeared to be required for its initiation. Labile LTP was not reversed by a PKC inhibitor and did not require either RIM1α or synaptotagmin-7, proteins implicated in other forms of presynaptic short-term plasticity. Similar NMDAR-independent potentiation could be elicited at synapses in mPFC. Labile LTP allows for rapid information storage that is erased under controlled circumstances and could have a role in a variety of hippocampal and prefrontal cortical computations related to short-term memory.SIGNIFICANCE STATEMENT Changes in synaptic strength are thought to represent information storage relevant to particular nervous system tasks. A single synapse can exhibit multiple overlapping forms of plasticity that shape information transfer from presynaptic to postsynaptic neurons. Here we investigate the mechanisms underlying labile LTP, an NMDAR-independent form of plasticity induced at hippocampal synapses. The potentiation is maintained for long periods as long as the synapses are infrequently active, but with regular activation, the synapses are depotentiated. Similar NMDAR-independent potentiation can also be induced at L2/3-to-L5 synapses in mPFC. Labile LTP requires a rise in presynaptic Ca2+ and protein kinase activation but is unaffected in RIM1α or synaptotagmin-7 mutant mice. Labile LTP may contribute to short-term or working memory in hippocampus and mPFC.


Asunto(s)
Hipocampo/fisiología , Potenciación a Largo Plazo/fisiología , Sinapsis/fisiología , Animales , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Plasticidad Neuronal/fisiología , Ratas , Ratas Sprague-Dawley
14.
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
15.
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
16.
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
17.
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
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...