Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Nature ; 624(7991): 317-332, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-38092916

RESUMEN

The mammalian brain consists of millions to billions of cells that are organized into many cell types with specific spatial distribution patterns and structural and functional properties1-3. Here we report a comprehensive and high-resolution transcriptomic and spatial cell-type atlas for the whole adult mouse brain. The cell-type atlas was created by combining a single-cell RNA-sequencing (scRNA-seq) dataset of around 7 million cells profiled (approximately 4.0 million cells passing quality control), and a spatial transcriptomic dataset of approximately 4.3 million cells using multiplexed error-robust fluorescence in situ hybridization (MERFISH). The atlas is hierarchically organized into 4 nested levels of classification: 34 classes, 338 subclasses, 1,201 supertypes and 5,322 clusters. We present an online platform, Allen Brain Cell Atlas, to visualize the mouse whole-brain cell-type atlas along with the single-cell RNA-sequencing and MERFISH datasets. We systematically analysed the neuronal and non-neuronal cell types across the brain and identified a high degree of correspondence between transcriptomic identity and spatial specificity for each cell type. The results reveal unique features of cell-type organization in different brain regions-in particular, a dichotomy between the dorsal and ventral parts of the brain. The dorsal part contains relatively fewer yet highly divergent neuronal types, whereas the ventral part contains more numerous neuronal types that are more closely related to each other. Our study also uncovered extraordinary diversity and heterogeneity in neurotransmitter and neuropeptide expression and co-expression patterns in different cell types. Finally, we found that transcription factors are major determinants of cell-type classification and identified a combinatorial transcription factor code that defines cell types across all parts of the brain. The whole mouse brain transcriptomic and spatial cell-type atlas establishes a benchmark reference atlas and a foundational resource for integrative investigations of cellular and circuit function, development and evolution of the mammalian brain.


Asunto(s)
Encéfalo , Perfilación de la Expresión Génica , Transcriptoma , Animales , Ratones , Encéfalo/anatomía & histología , Encéfalo/citología , Encéfalo/metabolismo , Conjuntos de Datos como Asunto , Hibridación Fluorescente in Situ , Vías Nerviosas , Neuronas/clasificación , Neuronas/metabolismo , Neuropéptidos/metabolismo , Neurotransmisores/metabolismo , ARN/análisis , Análisis de Expresión Génica de una Sola Célula , Factores de Transcripción/metabolismo , Transcriptoma/genética
2.
Science ; 382(6667): eade9516, 2023 10 13.
Artículo en Inglés | MEDLINE | ID: mdl-37824638

RESUMEN

The cognitive abilities of humans are distinctive among primates, but their molecular and cellular substrates are poorly understood. We used comparative single-nucleus transcriptomics to analyze samples of the middle temporal gyrus (MTG) from adult humans, chimpanzees, gorillas, rhesus macaques, and common marmosets to understand human-specific features of the neocortex. Human, chimpanzee, and gorilla MTG showed highly similar cell-type composition and laminar organization as well as a large shift in proportions of deep-layer intratelencephalic-projecting neurons compared with macaque and marmoset MTG. Microglia, astrocytes, and oligodendrocytes had more-divergent expression across species compared with neurons or oligodendrocyte precursor cells, and neuronal expression diverged more rapidly on the human lineage. Only a few hundred genes showed human-specific patterning, suggesting that relatively few cellular and molecular changes distinctively define adult human cortical structure.


Asunto(s)
Cognición , Hominidae , Neocórtex , Lóbulo Temporal , Animales , Humanos , Perfilación de la Expresión Génica , Gorilla gorilla/genética , Hominidae/genética , Hominidae/fisiología , Macaca mulatta/genética , Pan troglodytes/genética , Filogenia , Transcriptoma , Neocórtex/fisiología , Especificidad de la Especie , Lóbulo Temporal/fisiología
3.
Science ; 382(6667): eadf6812, 2023 10 13.
Artículo en Inglés | MEDLINE | ID: mdl-37824655

RESUMEN

Variation in cytoarchitecture is the basis for the histological definition of cortical areas. We used single cell transcriptomics and performed cellular characterization of the human cortex to better understand cortical areal specialization. Single-nucleus RNA-sequencing of 8 areas spanning cortical structural variation showed a highly consistent cellular makeup for 24 cell subclasses. However, proportions of excitatory neuron subclasses varied substantially, likely reflecting differences in connectivity across primary sensorimotor and association cortices. Laminar organization of astrocytes and oligodendrocytes also differed across areas. Primary visual cortex showed characteristic organization with major changes in the excitatory to inhibitory neuron ratio, expansion of layer 4 excitatory neurons, and specialized inhibitory neurons. These results lay the groundwork for a refined cellular and molecular characterization of human cortical cytoarchitecture and areal specialization.


Asunto(s)
Neocórtex , Humanos , Neocórtex/metabolismo , Neocórtex/ultraestructura , Neuronas/clasificación , Neuronas/metabolismo , Transcriptoma , Análisis de Expresión Génica de una Sola Célula , Filogenia
4.
Science ; 382(6667): eadf0805, 2023 10 13.
Artículo en Inglés | MEDLINE | ID: mdl-37824667

RESUMEN

Neocortical layer 1 (L1) is a site of convergence between pyramidal-neuron dendrites and feedback axons where local inhibitory signaling can profoundly shape cortical processing. Evolutionary expansion of human neocortex is marked by distinctive pyramidal neurons with extensive L1 branching, but whether L1 interneurons are similarly diverse is underexplored. Using Patch-seq recordings from human neurosurgical tissue, we identified four transcriptomic subclasses with mouse L1 homologs, along with distinct subtypes and types unmatched in mouse L1. Subclass and subtype comparisons showed stronger transcriptomic differences in human L1 and were correlated with strong morphoelectric variability along dimensions distinct from mouse L1 variability. Accompanied by greater layer thickness and other cytoarchitecture changes, these findings suggest that L1 has diverged in evolution, reflecting the demands of regulating the expanded human neocortical circuit.


Asunto(s)
Neocórtex , Animales , Humanos , Ratones , Axones/metabolismo , Interneuronas/metabolismo , Neocórtex/citología , Neocórtex/metabolismo , Células Piramidales/metabolismo , Transcriptoma
5.
Science ; 382(6667): eadf6484, 2023 10 13.
Artículo en Inglés | MEDLINE | ID: mdl-37824669

RESUMEN

Human cortex transcriptomic studies have revealed a hierarchical organization of γ-aminobutyric acid-producing (GABAergic) neurons from subclasses to a high diversity of more granular types. Rapid GABAergic neuron viral genetic labeling plus Patch-seq (patch-clamp electrophysiology plus single-cell RNA sequencing) sampling in human brain slices was used to reliably target and analyze GABAergic neuron subclasses and individual transcriptomic types. This characterization elucidated transitions between PVALB and SST subclasses, revealed morphological heterogeneity within an abundant transcriptomic type, identified multiple spatially distinct types of the primate-specialized double bouquet cells (DBCs), and shed light on cellular differences between homologous mouse and human neocortical GABAergic neuron types. These results highlight the importance of multimodal phenotypic characterization for refinement of emerging transcriptomic cell type taxonomies and for understanding conserved and specialized cellular properties of human brain cell types.


Asunto(s)
Neuronas GABAérgicas , Interneuronas , Neocórtex , Animales , Humanos , Ratones , Fenómenos Electrofisiológicos , Neuronas GABAérgicas/metabolismo , Ácido gamma-Aminobutírico/metabolismo , Interneuronas/metabolismo , Neocórtex/citología , Neocórtex/metabolismo , Técnicas de Placa-Clamp
6.
Neuron ; 111(22): 3590-3603.e5, 2023 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-37625400

RESUMEN

Although cardinal cortical interneuron identity is established upon cell-cycle exit, it remains unclear whether specific interneuron subtypes are pre-established, and if so, how their identity is maintained prior to circuit integration. We conditionally removed Sox6 (Sox6-cKO) in migrating somatostatin (Sst+) interneurons and assessed the effects on their mature identity. In adolescent mice, five of eight molecular Sst+ subtypes were nearly absent in the Sox6-cKO cortex without a reduction in cell number. Sox6-cKO cells displayed electrophysiological maturity and expressed genes enriched within the broad class of Sst+ interneurons. Furthermore, we could infer subtype identity prior to cortical integration (embryonic day 18.5), suggesting that the loss in subtype was due to disrupted subtype maintenance. Conversely, Sox6 removal at postnatal day 7 did not disrupt marker expression in the mature cortex. Therefore, Sox6 is necessary during migration for maintenance of Sst+ subtype identity, indicating that subtype maintenance requires active transcriptional programs.


Asunto(s)
Interneuronas , Somatostatina , Ratones , Animales , Interneuronas/fisiología , Somatostatina/metabolismo , Fenómenos Electrofisiológicos , Corteza Cerebral , Parvalbúminas/metabolismo
7.
bioRxiv ; 2023 Mar 06.
Artículo en Inglés | MEDLINE | ID: mdl-37034735

RESUMEN

The mammalian brain is composed of millions to billions of cells that are organized into numerous cell types with specific spatial distribution patterns and structural and functional properties. An essential step towards understanding brain function is to obtain a parts list, i.e., a catalog of cell types, of the brain. Here, we report a comprehensive and high-resolution transcriptomic and spatial cell type atlas for the whole adult mouse brain. The cell type atlas was created based on the combination of two single-cell-level, whole-brain-scale datasets: a single-cell RNA-sequencing (scRNA-seq) dataset of ~7 million cells profiled, and a spatially resolved transcriptomic dataset of ~4.3 million cells using MERFISH. The atlas is hierarchically organized into five nested levels of classification: 7 divisions, 32 classes, 306 subclasses, 1,045 supertypes and 5,200 clusters. We systematically analyzed the neuronal, non-neuronal, and immature neuronal cell types across the brain and identified a high degree of correspondence between transcriptomic identity and spatial specificity for each cell type. The results reveal unique features of cell type organization in different brain regions, in particular, a dichotomy between the dorsal and ventral parts of the brain: the dorsal part contains relatively fewer yet highly divergent neuronal types, whereas the ventral part contains more numerous neuronal types that are more closely related to each other. We also systematically characterized cell-type specific expression of neurotransmitters, neuropeptides, and transcription factors. The study uncovered extraordinary diversity and heterogeneity in neurotransmitter and neuropeptide expression and co-expression patterns in different cell types across the brain, suggesting they mediate a myriad of modes of intercellular communications. Finally, we found that transcription factors are major determinants of cell type classification in the adult mouse brain and identified a combinatorial transcription factor code that defines cell types across all parts of the brain. The whole-mouse-brain transcriptomic and spatial cell type atlas establishes a benchmark reference atlas and a foundational resource for deep and integrative investigations of cell type and circuit function, development, and evolution of the mammalian brain.

8.
Science ; 377(6601): 56-62, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35771910

RESUMEN

The human cerebral cortex has tremendous cellular diversity. How different cell types are organized in the human cortex and how cellular organization varies across species remain unclear. In this study, we performed spatially resolved single-cell profiling of 4000 genes using multiplexed error-robust fluorescence in situ hybridization (MERFISH), identified more than 100 transcriptionally distinct cell populations, and generated a molecularly defined and spatially resolved cell atlas of the human middle and superior temporal gyrus. We further explored cell-cell interactions arising from soma contact or proximity in a cell type-specific manner. Comparison of the human and mouse cortices showed conservation in the laminar organization of cells and differences in somatic interactions across species. Our data revealed human-specific cell-cell proximity patterns and a markedly increased enrichment for interactions between neurons and non-neuronal cells in the human cortex.


Asunto(s)
Corteza Cerebral , Perfilación de la Expresión Génica , Neuronas , Análisis de la Célula Individual , Animales , Comunicación Celular , Corteza Cerebral/citología , Corteza Cerebral/metabolismo , Humanos , Hibridación Fluorescente in Situ/métodos , Ratones , Neuronas/citología , Neuronas/metabolismo , Análisis de la Célula Individual/métodos
10.
Commun Biol ; 4(1): 998, 2021 08 24.
Artículo en Inglés | MEDLINE | ID: mdl-34429496

RESUMEN

The ability to spatially resolve the cellular architecture of human cortical cell types over informative areas is essential to understanding brain function. We combined in situ sequencing gene expression data and single-nucleus RNA-sequencing cell type definitions to spatially map cells in sections of the human cortex via probabilistic cell typing. We mapped and classified a total of 59,816 cells into all 75 previously defined subtypes to create a first spatial atlas of human cortical cells in their native position, their abundances and genetic signatures. We also examined the precise within- and across-layer distributions of all the cell types and provide a resource for the cell atlas community. The abundances and locations presented here could serve as a reference for further studies, that include human brain tissues and disease applications at the cell type level.


Asunto(s)
Corteza Cerebral/metabolismo , Mapeo Cromosómico , Transcriptoma , Adulto , Anciano , Humanos , Hibridación in Situ , Masculino , Análisis de Secuencia de ARN
11.
Nat Methods ; 18(8): 937-944, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34226720

RESUMEN

Fluorescence in situ hybridization (FISH) allows researchers to visualize the spatial position and quantity of nucleic acids in fixed samples. Recently, considerable progress has been made in developing oligonucleotide (oligo)-based FISH methods that have enabled researchers to study the three-dimensional organization of the genome at super-resolution and visualize the spatial patterns of gene expression for thousands of genes in individual cells. However, there are few existing computational tools to support the bioinformatics workflows necessary to carry out these experiments using oligo FISH probes. Here, we introduce paint server and homology optimization pipeline (PaintSHOP), an interactive platform for the design of oligo FISH experiments. PaintSHOP enables researchers to identify probes for their experimental targets efficiently, to incorporate additional necessary sequences such as primer pairs and to easily generate files documenting library design. PaintSHOP democratizes and standardizes the process of designing complex probe sets for the oligo FISH community.


Asunto(s)
Pintura Cromosómica/métodos , Biología Computacional/métodos , Genoma Humano , Hibridación Fluorescente in Situ/métodos , Sondas de Oligonucleótidos/química , Secuencias Repetitivas de Ácidos Nucleicos , Transcriptoma , Humanos
13.
Nature ; 573(7772): 61-68, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31435019

RESUMEN

Elucidating the cellular architecture of the human cerebral cortex is central to understanding our cognitive abilities and susceptibility to disease. Here we used single-nucleus RNA-sequencing analysis to perform a comprehensive study of cell types in the middle temporal gyrus of human cortex. We identified a highly diverse set of excitatory and inhibitory neuron types that are mostly sparse, with excitatory types being less layer-restricted than expected. Comparison to similar mouse cortex single-cell RNA-sequencing datasets revealed a surprisingly well-conserved cellular architecture that enables matching of homologous types and predictions of properties of human cell types. Despite this general conservation, we also found extensive differences between homologous human and mouse cell types, including marked alterations in proportions, laminar distributions, gene expression and morphology. These species-specific features emphasize the importance of directly studying human brain.


Asunto(s)
Astrocitos/clasificación , Evolución Biológica , Corteza Cerebral/citología , Corteza Cerebral/metabolismo , Neuronas/clasificación , Adolescente , Adulto , Anciano , Animales , Astrocitos/citología , Femenino , Humanos , Masculino , Ratones , Persona de Mediana Edad , Inhibición Neural , Neuronas/citología , Análisis de Componente Principal , RNA-Seq , Análisis de la Célula Individual , Especificidad de la Especie , Transcriptoma/genética , Adulto Joven
14.
Neuron ; 100(5): 1194-1208.e5, 2018 12 05.
Artículo en Inglés | MEDLINE | ID: mdl-30392798

RESUMEN

Gene expression studies suggest that differential ion channel expression contributes to differences in rodent versus human neuronal physiology. We tested whether h-channels more prominently contribute to the physiological properties of human compared to mouse supragranular pyramidal neurons. Single-cell/nucleus RNA sequencing revealed ubiquitous HCN1-subunit expression in excitatory neurons in human, but not mouse, supragranular layers. Using patch-clamp recordings, we found stronger h-channel-related membrane properties in supragranular pyramidal neurons in human temporal cortex, compared to mouse supragranular pyramidal neurons in temporal association area. The magnitude of these differences depended upon cortical depth and was largest in pyramidal neurons in deep L3. Additionally, pharmacologically blocking h-channels produced a larger change in membrane properties in human compared to mouse neurons. Finally, using biophysical modeling, we provide evidence that h-channels promote the transfer of theta frequencies from dendrite-to-soma in human L3 pyramidal neurons. Thus, h-channels contribute to between-species differences in a fundamental neuronal property.


Asunto(s)
Corteza Cerebral/fisiología , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/fisiología , Potenciales de la Membrana , Canales de Potasio/fisiología , Células Piramidales/fisiología , Adulto , Animales , Membrana Celular/fisiología , Corteza Cerebral/metabolismo , Femenino , Humanos , Canales Regulados por Nucleótidos Cíclicos Activados por Hiperpolarización/metabolismo , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , Modelos Neurológicos , Canales de Potasio/metabolismo , Células Piramidales/metabolismo , Especificidad de la Especie
15.
Nat Neurosci ; 21(9): 1185-1195, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-30150662

RESUMEN

We describe convergent evidence from transcriptomics, morphology, and physiology for a specialized GABAergic neuron subtype in human cortex. Using unbiased single-nucleus RNA sequencing, we identify ten GABAergic interneuron subtypes with combinatorial gene signatures in human cortical layer 1 and characterize a group of human interneurons with anatomical features never described in rodents, having large 'rosehip'-like axonal boutons and compact arborization. These rosehip cells show an immunohistochemical profile (GAD1+CCK+, CNR1-SST-CALB2-PVALB-) matching a single transcriptomically defined cell type whose specific molecular marker signature is not seen in mouse cortex. Rosehip cells in layer 1 make homotypic gap junctions, predominantly target apical dendritic shafts of layer 3 pyramidal neurons, and inhibit backpropagating pyramidal action potentials in microdomains of the dendritic tuft. These cells are therefore positioned for potent local control of distal dendritic computation in cortical pyramidal neurons.


Asunto(s)
Corteza Cerebral/metabolismo , Corteza Cerebral/ultraestructura , Neuronas GABAérgicas/metabolismo , Neuronas GABAérgicas/ultraestructura , Transcriptoma , Adulto , Anciano , Axones/ultraestructura , Espinas Dendríticas/metabolismo , Espinas Dendríticas/ultraestructura , Uniones Comunicantes/metabolismo , Uniones Comunicantes/ultraestructura , Biblioteca de Genes , Humanos , Masculino , Reacción en Cadena de la Polimerasa , Terminales Presinápticos/metabolismo , Terminales Presinápticos/ultraestructura , Células Piramidales/metabolismo , Células Piramidales/ultraestructura , ARN/análisis , ARN/genética , Análisis de Secuencia de ARN
17.
Cell Stem Cell ; 21(3): 289-290, 2017 09 07.
Artículo en Inglés | MEDLINE | ID: mdl-28886360

RESUMEN

3D organoids enable in vitro human brain development models, but they have not yet recapitulated some essential features of brain circuit formation. Recently, several studies appearing in Nature, Nature Methods, and Cell Stem Cell generated fused organoid models of inhibitory and excitatory neuron development, which can now achieve functional circuit integration.


Asunto(s)
Neurogénesis , Organoides , Encéfalo , Movimiento Celular , Humanos , Interneuronas
18.
Neuron ; 93(5): 1035-1048.e5, 2017 Mar 08.
Artículo en Inglés | MEDLINE | ID: mdl-28279351

RESUMEN

GABAergic interneurons are essential for neural circuit function, and their loss or dysfunction is implicated in human neuropsychiatric disease. In vitro methods for interneuron generation hold promise for studying human cellular and functional properties and, ultimately, for therapeutic cell replacement. Here we describe a protocol for generating cortical interneurons from hESCs and analyze the properties and maturation time course of cell types using single-cell RNA-seq. We find that the cell types produced mimic in vivo temporal patterns of neuron and glial production, with immature progenitors and neurons observed early and mature cortical neurons and glial cell types produced late. By comparing the transcriptomes of immature interneurons to those of more mature neurons, we identified genes important for human interneuron differentiation. Many of these genes were previously implicated in neurodevelopmental and neuropsychiatric disorders.


Asunto(s)
Diferenciación Celular/fisiología , Movimiento Celular/fisiología , Neuronas GABAérgicas/citología , Interneuronas/citología , Proteínas del Tejido Nervioso/metabolismo , Neuroglía/citología , Células Cultivadas , Humanos , Neurogénesis/fisiología , Análisis de la Célula Individual , Factores de Transcripción/metabolismo
19.
J Neurosci ; 32(49): 17690-705, 2012 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-23223290

RESUMEN

Although previous work identified transcription factors crucial for the specification and migration of parvalbumin (PV)-expressing and somatostatin (SST)-expressing interneurons, the intrinsic factors required for the terminal differentiation, connectivity, and survival of these cell types remain uncharacterized. Here we demonstrate that, within subpopulations of cortical interneurons, Satb1 (special AT-rich binding protein) promotes terminal differentiation, connectivity, and survival in interneurons that express PV and SST. We find that conditional removal of Satb1 in mouse interneurons results in the loss of a majority of SST-expressing cells across all cortical layers, as well as some PV-expressing cells in layers IV and VI, by postnatal day 21. SST-expressing cells initially migrate to the cortex in Satb1 mutant mice, but receive reduced levels of afferent input and begin to die during the first postnatal week. Electrophysiological characterization indicates that loss of Satb1 function in interneurons results in a loss of functional inhibition of excitatory principal cells. These data suggest that Satb1 is required for medial ganglionic eminence-derived interneuron differentiation, connectivity, and survival.


Asunto(s)
Diferenciación Celular/fisiología , Corteza Cerebral/crecimiento & desarrollo , Interneuronas/fisiología , Proteínas de Unión a la Región de Fijación a la Matriz/fisiología , Terminales Presinápticos/fisiología , Animales , Ondas Encefálicas/fisiología , Movimiento Celular/fisiología , Supervivencia Celular/fisiología , Corteza Cerebral/citología , Corteza Cerebral/metabolismo , Corteza Cerebral/fisiología , Potenciales Postsinápticos Excitadores/fisiología , Regulación del Desarrollo de la Expresión Génica/fisiología , Interneuronas/citología , Interneuronas/metabolismo , Proteínas de Unión a la Región de Fijación a la Matriz/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Inhibición Neural/fisiología , Parvalbúminas/metabolismo , Terminales Presinápticos/metabolismo , Somatostatina/metabolismo , Factores de Transcripción/metabolismo , Factores de Transcripción/fisiología
20.
J Neurosci ; 28(15): 3966-75, 2008 Apr 09.
Artículo en Inglés | MEDLINE | ID: mdl-18400896

RESUMEN

Olfactory bulb (OB) interneurons are a heterogeneous population produced beginning in embryogenesis and continuing through adulthood. Understanding how this diversity arises will provide insight into how OB microcircuitry is established as well as adult neurogenesis. Particular spatial domains have been shown to contribute specific interneuron subtypes. However, the temporal profile by which OB interneuron subtypes are produced is unknown. Using inducible genetic fate mapping of Dlx1/2 precursors, we analyzed the production of seven OB interneuron subtypes and found that the generation of each subpopulation has a unique temporal signature. Within the glomerular layer, the production of tyrosine hydroxylase-positive interneurons is maximal during early embryogenesis and decreases thereafter. In contrast, the generation of calbindin interneurons is maximal during late embryogenesis and declines postnatally, whereas calretinin (CR) cell production is low during embryogenesis and increases postnatally. Parvalbumin interneurons within the external plexiform layer are produced only perinatally, whereas the generation of 5T4-positive granule cells in the mitral cell layer does not change significantly over time. CR-positive granule cells are not produced at early embryonic time points, but constitute a large percentage of the granule cells born after birth. Blanes cells in contrast are produced in greatest number during embryogenesis. Together we provide the first comprehensive analysis of the temporal generation of OB interneuron subtypes and demonstrate that the timing by which these populations are produced is tightly orchestrated.


Asunto(s)
Diferenciación Celular , Interneuronas/clasificación , Interneuronas/citología , Bulbo Olfatorio/citología , Envejecimiento/fisiología , Animales , Animales Recién Nacidos , Embrión de Mamíferos/citología , Desarrollo Embrionario , Femenino , Interneuronas/metabolismo , Masculino , Ratones , Ratones Transgénicos , Bulbo Olfatorio/embriología , Bulbo Olfatorio/crecimiento & desarrollo , Bulbo Olfatorio/metabolismo , Células Madre/citología , Células Madre/metabolismo , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...