Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 1.353
Filtrar
Más filtros

Tipo del documento
Publication year range
1.
Annu Rev Immunol ; 39: 313-344, 2021 04 26.
Artículo en Inglés | MEDLINE | ID: mdl-33902313

RESUMEN

Tissue-resident macrophages are present in most tissues with developmental, self-renewal, or functional attributes that do not easily fit into a textbook picture of a plastic and multifunctional macrophage originating from hematopoietic stem cells; nor does it fit a pro- versus anti-inflammatory paradigm. This review presents and discusses current knowledge on the developmental biology of macrophages from an evolutionary perspective focused on the function of macrophages, which may aid in study of developmental, inflammatory, tumoral, and degenerative diseases. We also propose a framework to investigate the functions of macrophages in vivo and discuss how inherited germline and somatic mutations may contribute to the roles of macrophages in diseases.


Asunto(s)
Células Madre Hematopoyéticas , Macrófagos , Animales , Biología , Humanos
2.
Cell ; 187(11): 2855-2874.e19, 2024 May 23.
Artículo en Inglés | MEDLINE | ID: mdl-38657603

RESUMEN

Progress in understanding early human development has been impeded by the scarcity of reference datasets from natural embryos, particularly those with spatial information during crucial stages like gastrulation. We conducted high-resolution spatial transcriptomics profiling on 38,562 spots from 62 transverse sections of an intact Carnegie stage (CS) 8 human embryo. From this spatial transcriptomic dataset, we constructed a 3D model of the CS8 embryo, in which a range of cell subtypes are identified, based on gene expression patterns and positional register, along the anterior-posterior, medial-lateral, and dorsal-ventral axis in the embryo. We further characterized the lineage trajectories of embryonic and extra-embryonic tissues and associated regulons and the regionalization of signaling centers and signaling activities that underpin lineage progression and tissue patterning during gastrulation. Collectively, the findings of this study provide insights into gastrulation and post-gastrulation development of the human embryo.


Asunto(s)
Embrión de Mamíferos , Gastrulación , Regulación del Desarrollo de la Expresión Génica , Imagenología Tridimensional , Humanos , Embrión de Mamíferos/metabolismo , Transcriptoma/genética , Gástrula/metabolismo , Gástrula/embriología , Transducción de Señal , Linaje de la Célula , Perfilación de la Expresión Génica , Tipificación del Cuerpo/genética
3.
Cell ; 187(12): 3141-3160.e23, 2024 Jun 06.
Artículo en Inglés | MEDLINE | ID: mdl-38759650

RESUMEN

Systematic functional profiling of the gene set that directs embryonic development is an important challenge. To tackle this challenge, we used 4D imaging of C. elegans embryogenesis to capture the effects of 500 gene knockdowns and developed an automated approach to compare developmental phenotypes. The automated approach quantifies features-including germ layer cell numbers, tissue position, and tissue shape-to generate temporal curves whose parameterization yields numerical phenotypic signatures. In conjunction with a new similarity metric that operates across phenotypic space, these signatures enabled the generation of ranked lists of genes predicted to have similar functions, accessible in the PhenoBank web portal, for ∼25% of essential development genes. The approach identified new gene and pathway relationships in cell fate specification and morphogenesis and highlighted the utilization of specialized energy generation pathways during embryogenesis. Collectively, the effort establishes the foundation for comprehensive analysis of the gene set that builds a multicellular organism.


Asunto(s)
Caenorhabditis elegans , Desarrollo Embrionario , Regulación del Desarrollo de la Expresión Génica , Animales , Caenorhabditis elegans/embriología , Caenorhabditis elegans/genética , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/genética , Embrión no Mamífero/metabolismo , Perfilación de la Expresión Génica/métodos , Técnicas de Silenciamiento del Gen , Fenotipo
4.
Cell ; 186(12): 2610-2627.e18, 2023 06 08.
Artículo en Inglés | MEDLINE | ID: mdl-37209682

RESUMEN

The hourglass model describes the convergence of species within the same phylum to a similar body plan during development; however, the molecular mechanisms underlying this phenomenon in mammals remain poorly described. Here, we compare rabbit and mouse time-resolved differentiation trajectories to revisit this model at single-cell resolution. We modeled gastrulation dynamics using hundreds of embryos sampled between gestation days 6.0 and 8.5 and compared the species using a framework for time-resolved single-cell differentiation-flows analysis. We find convergence toward similar cell-state compositions at E7.5, supported by the quantitatively conserved expression of 76 transcription factors, despite divergence in surrounding trophoblast and hypoblast signaling. However, we observed noticeable changes in specification timing of some lineages and divergence of primordial germ cell programs, which in the rabbit do not activate mesoderm genes. Comparative analysis of temporal differentiation models provides a basis for studying the evolution of gastrulation dynamics across mammals.


Asunto(s)
Gastrulación , Mesodermo , Animales , Conejos , Ratones , Gastrulación/genética , Mesodermo/fisiología , Diferenciación Celular/fisiología , Mamíferos/genética , Trofoblastos , Regulación del Desarrollo de la Expresión Génica
5.
Cell ; 181(4): 922-935.e21, 2020 05 14.
Artículo en Inglés | MEDLINE | ID: mdl-32315617

RESUMEN

Single-cell RNA sequencing (scRNA-seq) provides a leap forward in resolving cellular diversity and developmental trajectories but fails to comprehensively delineate the spatial organization and precise cellular makeup of individual embryos. Here, we reconstruct from scRNA-seq and light sheet imaging data a canonical digital embryo that captures the genome-wide gene expression trajectory of every single cell at every cell division in the 18 lineages up to gastrulation in the ascidian Phallusia mammillata. By using high-coverage scRNA-seq, we devise a computational framework that stratifies single cells of individual embryos into cell types without prior knowledge. Unbiased transcriptome data analysis mapped each cell's physical position and lineage history, yielding the complete history of gene expression at the genome-wide level for every single cell in a developing embryo. A comparison of individual embryos reveals both extensive reproducibility between symmetric embryo sides and a large inter-embryonic variability due to small differences in embryogenesis timing.


Asunto(s)
Perfilación de la Expresión Génica/métodos , Análisis de Secuencia de ARN/métodos , Análisis de la Célula Individual/métodos , Animales , Linaje de la Célula/genética , Cordados/genética , Biología Computacional/métodos , Gastrulación/genética , Regulación del Desarrollo de la Expresión Génica/genética , Reproducibilidad de los Resultados , Transcriptoma/genética , Urocordados/genética
6.
Cell ; 176(6): 1379-1392.e14, 2019 03 07.
Artículo en Inglés | MEDLINE | ID: mdl-30773315

RESUMEN

Cell fate specification by lateral inhibition typically involves contact signaling through the Delta-Notch signaling pathway. However, whether this is the only signaling mode mediating lateral inhibition remains unclear. Here we show that in zebrafish oogenesis, a group of cells within the granulosa cell layer at the oocyte animal pole acquire elevated levels of the transcriptional coactivator TAZ in their nuclei. One of these cells, the future micropyle precursor cell (MPC), accumulates increasingly high levels of nuclear TAZ and grows faster than its surrounding cells, mechanically compressing those cells, which ultimately lose TAZ from their nuclei. Strikingly, relieving neighbor-cell compression by MPC ablation or aspiration restores nuclear TAZ accumulation in neighboring cells, eventually leading to MPC re-specification from these cells. Conversely, MPC specification is defective in taz-/- follicles. These findings uncover a novel mode of lateral inhibition in cell fate specification based on mechanical signals controlling TAZ activity.


Asunto(s)
Péptidos y Proteínas de Señalización Intracelular/metabolismo , Oogénesis/fisiología , Proteínas de Pez Cebra/metabolismo , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Comunicación Celular/fisiología , Diferenciación Celular/fisiología , Linaje de la Célula , Núcleo Celular/metabolismo , Femenino , Células de la Granulosa/metabolismo , Péptidos y Proteínas de Señalización Intracelular/antagonistas & inhibidores , Oocitos/metabolismo , Oocitos/fisiología , Proteínas Serina-Treonina Quinasas/metabolismo , Transducción de Señal , Factores de Transcripción/metabolismo , Activación Transcripcional/fisiología , Proteínas Coactivadoras Transcripcionales con Motivo de Unión a PDZ , Pez Cebra/metabolismo , Proteínas de Pez Cebra/antagonistas & inhibidores
7.
Cell ; 176(4): 844-855.e15, 2019 02 07.
Artículo en Inglés | MEDLINE | ID: mdl-30712870

RESUMEN

In developing organisms, spatially prescribed cell identities are thought to be determined by the expression levels of multiple genes. Quantitative tests of this idea, however, require a theoretical framework capable of exposing the rules and precision of cell specification over developmental time. We use the gap gene network in the early fly embryo as an example to show how expression levels of the four gap genes can be jointly decoded into an optimal specification of position with 1% accuracy. The decoder correctly predicts, with no free parameters, the dynamics of pair-rule expression patterns at different developmental time points and in various mutant backgrounds. Precise cellular identities are thus available at the earliest stages of development, contrasting the prevailing view of positional information being slowly refined across successive layers of the patterning network. Our results suggest that developmental enhancers closely approximate a mathematically optimal decoding strategy.


Asunto(s)
Proteínas Activadoras de GTPasa/genética , Regulación del Desarrollo de la Expresión Génica/genética , Redes Reguladoras de Genes/genética , Animales , Tipificación del Cuerpo/genética , Diferenciación Celular/genética , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Embrión no Mamífero/metabolismo , Desarrollo Embrionario/genética , Proteínas Activadoras de GTPasa/metabolismo , Regulación del Desarrollo de la Expresión Génica/fisiología , Modelos Genéticos , Factores de Transcripción/metabolismo
8.
Cell ; 171(3): 573-587.e14, 2017 Oct 19.
Artículo en Inglés | MEDLINE | ID: mdl-29033129

RESUMEN

Progenitor cells differentiate into specialized cell types through coordinated expression of lineage-specific genes and modification of complex chromatin configurations. We demonstrate that a histone deacetylase (Hdac3) organizes heterochromatin at the nuclear lamina during cardiac progenitor lineage restriction. Specification of cardiomyocytes is associated with reorganization of peripheral heterochromatin, and independent of deacetylase activity, Hdac3 tethers peripheral heterochromatin containing lineage-relevant genes to the nuclear lamina. Deletion of Hdac3 in cardiac progenitor cells releases genomic regions from the nuclear periphery, leading to precocious cardiac gene expression and differentiation into cardiomyocytes; in contrast, restricting Hdac3 to the nuclear periphery rescues myogenesis in progenitors otherwise lacking Hdac3. Our results suggest that availability of genomic regions for activation by lineage-specific factors is regulated in part through dynamic chromatin-nuclear lamina interactions and that competence of a progenitor cell to respond to differentiation signals may depend upon coordinated movement of responding gene loci away from the nuclear periphery.


Asunto(s)
Cromatina/metabolismo , Regulación del Desarrollo de la Expresión Génica , Histona Desacetilasas/metabolismo , Lámina Nuclear/metabolismo , Células Madre/citología , Animales , Genoma , Ratones , Miocitos Cardíacos/citología , Miocitos Cardíacos/metabolismo , Células Madre/metabolismo
9.
Annu Rev Cell Dev Biol ; 34: 451-469, 2018 10 06.
Artículo en Inglés | MEDLINE | ID: mdl-30028642

RESUMEN

Posttranscriptional mechanisms provide powerful means to expand the coding power of genomes. In nervous systems, alternative splicing has emerged as a fundamental mechanism not only for the diversification of protein isoforms but also for the spatiotemporal control of transcripts. Thus, alternative splicing programs play instructive roles in the development of neuronal cell type-specific properties, neuronal growth, self-recognition, synapse specification, and neuronal network function. Here we discuss the most recent genome-wide efforts on mapping RNA codes and RNA-binding proteins for neuronal alternative splicing regulation. We illustrate how alternative splicing shapes key steps of neuronal development, neuronal maturation, and synaptic properties. Finally, we highlight efforts to dissect the spatiotemporal dynamics of alternative splicing and their potential contribution to neuronal plasticity and the mature nervous system.


Asunto(s)
Empalme Alternativo/genética , Diferenciación Celular/genética , Plasticidad Neuronal/genética , Neuronas/metabolismo , Humanos , Neurogénesis/genética , Neuronas/citología , Isoformas de Proteínas/genética , ARN/genética , Proteínas de Unión al ARN/genética , Sinapsis/genética
10.
Annu Rev Cell Dev Biol ; 32: 77-101, 2016 10 06.
Artículo en Inglés | MEDLINE | ID: mdl-27501446

RESUMEN

Tissue-specific transcription regulators emerged as key developmental control genes, which operate in the context of complex gene regulatory networks (GRNs) to coordinate progressive cell fate specification and tissue morphogenesis. We discuss how GRNs control the individual cell behaviors underlying complex morphogenetic events. Cell behaviors classically range from mesenchymal cell motility to cell shape changes in epithelial sheets. These behaviors emerge from the tissue-specific, multiscale integration of the local activities of universal and pleiotropic effectors, which underlie modular subcellular processes including cytoskeletal dynamics, cell-cell and cell-matrix adhesion, signaling, polarity, and vesicle trafficking. Extrinsic cues and intrinsic cell competence determine the subcellular spatiotemporal patterns of effector activities. GRNs influence most subcellular activities by controlling only a fraction of the effector-coding genes, which we argue is enriched in effectors involved in reading and processing the extrinsic cues to contextualize intrinsic subcellular processes and canalize developmental cell behaviors. The properties of the transcription-cell behavior interface have profound implications for evolution and disease.


Asunto(s)
Células/metabolismo , Regulación del Desarrollo de la Expresión Génica , Transcripción Genética , Animales , Redes Reguladoras de Genes , Genómica , Humanos , Especificidad de Órganos/genética
11.
Annu Rev Cell Dev Biol ; 32: 47-75, 2016 10 06.
Artículo en Inglés | MEDLINE | ID: mdl-27576120

RESUMEN

Land plants can grow to tremendous body sizes, yet even the most complex architectures are the result of iterations of the same developmental processes: organ initiation, growth, and pattern formation. A central question in plant biology is how these processes are regulated and coordinated to allow for the formation of ordered, 3D structures. All these elementary processes first occur in early embryogenesis, during which, from a fertilized egg cell, precursors for all major tissues and stem cells are initiated, followed by tissue growth and patterning. Here we discuss recent progress in our understanding of this phase of plant life. We consider the cellular basis for multicellular development in 3D and focus on the genetic regulatory mechanisms that direct specific steps during early embryogenesis.


Asunto(s)
Morfogénesis , Semillas/embriología , Tipificación del Cuerpo , Nicho de Células Madre
12.
Annu Rev Genet ; 55: 349-376, 2021 11 23.
Artículo en Inglés | MEDLINE | ID: mdl-34546797

RESUMEN

Neural crest stem/progenitor cells arise early during vertebrate embryogenesis at the border of the forming central nervous system. They subsequently migrate throughout the body, eventually differentiating into diverse cell types ranging from neurons and glia of the peripheral nervous system to bones of the face, portions of the heart, and pigmentation of the skin. Along the body axis, the neural crest is heterogeneous, with different subpopulations arising in the head, neck, trunk, and tail regions, each characterized by distinct migratory patterns and developmental potential. Modern genomic approaches like single-cell RNA- and ATAC-sequencing (seq) have greatly enhanced our understanding of cell lineage trajectories and gene regulatory circuitry underlying the developmental progression of neural crest cells. Here, we discuss how genomic approaches have provided new insights into old questions in neural crest biology by elucidating transcriptional and posttranscriptional mechanisms that govern neural crest formation and the establishment of axial level identity.


Asunto(s)
Cresta Neural , Neuronas , Diferenciación Celular/genética , Linaje de la Célula/genética , Movimiento Celular/genética , Desarrollo Embrionario , Regulación del Desarrollo de la Expresión Génica/genética , Genómica , Neuronas/metabolismo
13.
Immunity ; 52(6): 1105-1118.e9, 2020 06 16.
Artículo en Inglés | MEDLINE | ID: mdl-32553173

RESUMEN

The challenges in recapitulating in vivo human T cell development in laboratory models have posed a barrier to understanding human thymopoiesis. Here, we used single-cell RNA sequencing (sRNA-seq) to interrogate the rare CD34+ progenitor and the more differentiated CD34- fractions in the human postnatal thymus. CD34+ thymic progenitors were comprised of a spectrum of specification and commitment states characterized by multilineage priming followed by gradual T cell commitment. The earliest progenitors in the differentiation trajectory were CD7- and expressed a stem-cell-like transcriptional profile, but had also initiated T cell priming. Clustering analysis identified a CD34+ subpopulation primed for the plasmacytoid dendritic lineage, suggesting an intrathymic dendritic specification pathway. CD2 expression defined T cell commitment stages where loss of B cell potential preceded that of myeloid potential. These datasets delineate gene expression profiles spanning key differentiation events in human thymopoiesis and provide a resource for the further study of human T cell development.


Asunto(s)
Diferenciación Celular/genética , Linaje de la Célula/genética , Linfopoyesis/genética , Linfocitos T/metabolismo , Timocitos/metabolismo , Animales , Biomarcadores , Biología Computacional , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Humanos , Inmunofenotipificación , Ratones , Análisis de la Célula Individual , Linfocitos T/citología , Timocitos/citología , Transcriptoma
14.
EMBO J ; 43(12): 2308-2336, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38760574

RESUMEN

How cells coordinate morphogenetic cues and fate specification during development remains a fundamental question in organogenesis. The mammary gland arises from multipotent stem cells (MaSCs), which are progressively replaced by unipotent progenitors by birth. However, the lack of specific markers for early fate specification has prevented the delineation of the features and spatial localization of MaSC-derived lineage-committed progenitors. Here, using single-cell RNA sequencing from E13.5 to birth, we produced an atlas of matched mouse mammary epithelium and mesenchyme and reconstructed the differentiation trajectories of MaSCs toward basal and luminal fate. We show that murine MaSCs exhibit lineage commitment just prior to the first sprouting events of mammary branching morphogenesis at E15.5. We identify early molecular markers for committed and multipotent MaSCs and define their spatial distribution within the developing tissue. Furthermore, we show that the mammary embryonic mesenchyme is composed of two spatially restricted cell populations, and that dermal mesenchyme-produced FGF10 is essential for embryonic mammary branching morphogenesis. Altogether, our data elucidate the spatiotemporal signals underlying lineage specification of multipotent MaSCs, and uncover the signals from mesenchymal cells that guide mammary branching morphogenesis.


Asunto(s)
Linaje de la Célula , Células Epiteliales , Glándulas Mamarias Animales , Células Madre Mesenquimatosas , Animales , Ratones , Glándulas Mamarias Animales/citología , Glándulas Mamarias Animales/embriología , Glándulas Mamarias Animales/metabolismo , Femenino , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Células Epiteliales/citología , Células Epiteliales/metabolismo , Diferenciación Celular , Células Madre Multipotentes/citología , Células Madre Multipotentes/metabolismo , Factor 10 de Crecimiento de Fibroblastos/metabolismo , Factor 10 de Crecimiento de Fibroblastos/genética , Morfogénesis , Análisis de la Célula Individual , Mesodermo/citología , Mesodermo/metabolismo , Mesodermo/embriología
15.
Annu Rev Neurosci ; 43: 1-30, 2020 07 08.
Artículo en Inglés | MEDLINE | ID: mdl-31299170

RESUMEN

Cortical interneurons display striking differences in shape, physiology, and other attributes, challenging us to appropriately classify them. We previously suggested that interneuron types should be defined by their role in cortical processing. Here, we revisit the question of how to codify their diversity based upon their division of labor and function as controllers of cortical information flow. We suggest that developmental trajectories provide a guide for appreciating interneuron diversity and argue that subtype identity is generated using a configurational (rather than combinatorial) code of transcription factors that produce attractor states in the underlying gene regulatory network. We present our updated three-stage model for interneuron specification: an initial cardinal step, allocating interneurons into a few major classes, followed by definitive refinement, creating subclasses upon settling within the cortex, and lastly, state determination, reflecting the incorporation of interneurons into functional circuit ensembles. We close by discussing findings indicating that major interneuron classes are both evolutionarily ancient and conserved. We propose that the complexity of cortical circuits is generated by phylogenetically old interneuron types, complemented by an evolutionary increase in principal neuron diversity. This suggests that a natural neurobiological definition of interneuron types might be derived from a match between their developmental origin and computational function.


Asunto(s)
Diferenciación Celular/fisiología , Corteza Cerebral/fisiología , Interneuronas/fisiología , Neurogénesis/fisiología , Animales , Humanos , Neuronas/fisiología , Factores de Transcripción/metabolismo
16.
Genes Dev ; 34(9-10): 621-636, 2020 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-32241803

RESUMEN

Peripheral somatosensory input is modulated in the dorsal spinal cord by a network of excitatory and inhibitory interneurons. PTF1A is a transcription factor essential in dorsal neural tube progenitors for specification of these inhibitory neurons. Thus, mechanisms regulating Ptf1a expression are key for generating neuronal circuits underlying somatosensory behaviors. Mutations targeted to distinct cis-regulatory elements for Ptf1a in mice, tested the in vivo contribution of each element individually and in combination. Mutations in an autoregulatory enhancer resulted in reduced levels of PTF1A, and reduced numbers of specific dorsal spinal cord inhibitory neurons, particularly those expressing Pdyn and Gal Although these mutants survive postnatally, at ∼3-5 wk they elicit a severe scratching phenotype. Behaviorally, the mutants have increased sensitivity to itch, but acute sensitivity to other sensory stimuli such as mechanical or thermal pain is unaffected. We demonstrate a requirement for positive transcriptional autoregulatory feedback to attain the level of the neuronal specification factor PTF1A necessary for generating correctly balanced neuronal circuits.


Asunto(s)
Retroalimentación Fisiológica/fisiología , Regulación de la Expresión Génica/fisiología , Neuronas/fisiología , Prurito/genética , Factores de Transcripción/genética , Animales , Sistemas CRISPR-Cas , Elementos de Facilitación Genéticos/genética , Ratones , Mutación , Neuronas/citología , Médula Espinal , Factores de Transcripción/metabolismo
17.
Development ; 151(3)2024 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-38205939

RESUMEN

Asymmetric cell divisions often generate daughter cells of unequal size in addition to different fates. In some contexts, daughter cell size asymmetry is thought to be a key input to specific binary cell fate decisions. An alternative possibility is that unequal division is a mechanism by which a variety of cells of different sizes are generated during embryonic development. We show here that two unequal cell divisions precede neuroblast formation in the C lineage of Caenorhabditis elegans. The equalisation of these divisions in a pig-1/MELK mutant background has little effect on neuroblast specification. Instead, we demonstrate that let-19/MDT13 is a regulator of the proneural basic helix-loop-helix transcription factor hlh-14/ASCL1 and find that both are required to concomitantly regulate the acquisition of neuroblast identity and neuroblast cell size. Thus, embryonic neuroblast cell size in this lineage is progressively regulated in parallel with identity by key neural cell fate regulators. We propose that key cell fate determinants have a previously unappreciated function in regulating unequal cleavage, and therefore cell size, of the progenitor cells whose daughter cell fates they then go on to specify.


Asunto(s)
Proteínas de Caenorhabditis elegans , Células-Madre Neurales , Animales , Proteínas de Caenorhabditis elegans/genética , Neuronas , Caenorhabditis elegans , División Celular , Tamaño de la Célula
18.
Development ; 151(3)2024 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-38230563

RESUMEN

An unanswered question in neurobiology is how are diverse neuron cell types generated from a small number of neural stem cells? In the Drosophila larval central brain, there are eight bilateral Type 2 neuroblast (T2NB) lineages that express a suite of early temporal factors followed by a different set of late temporal factors and generate the majority of the central complex (CX) neurons. The early-to-late switch is triggered by the orphan nuclear hormone receptor Seven-up (Svp), yet little is known about how this Svp-dependent switch is involved in specifying CX neuron identities. Here, we: (1) birth date the CX neurons P-EN and P-FN (early and late, respectively); (2) show that Svp is transiently expressed in all early T2NBs; and (3) show that loss of Svp expands the population of early born P-EN neurons at the expense of late born P-FN neurons. Furthermore, in the absence of Svp, T2NBs fail decommissioning and abnormally extend their lineage into week-old adults. We conclude that Svp is required to specify CX neuron identity, as well as to initiate T2NB decommissioning.


Asunto(s)
Proteínas de Drosophila , Células-Madre Neurales , Animales , Factores de Transcripción/metabolismo , Proteínas de Unión al ADN/metabolismo , Proteínas de Drosophila/metabolismo , Células-Madre Neurales/metabolismo , Neuronas/metabolismo , Drosophila/metabolismo , Linaje de la Célula/fisiología , Drosophila melanogaster/metabolismo
19.
Development ; 2024 Jul 22.
Artículo en Inglés | MEDLINE | ID: mdl-39037017

RESUMEN

Following up on our previous observation that early B cell factor (EBF) sites are enriched in open chromatin of the developing sensory epithelium of the mouse cochlea, we investigated the effect of deletion of Ebf1 on inner ear development. We used a Cre driver to delete Ebf1 at the otocyst stage prior to development of the cochlea. We examined the cochlea at postnatal day (P) 1 and found that the sensory epithelium had doubled in size but the length of the cochlear duct was unaffected. We also found that deletion of Ebf1 led to ectopic sensory patches in the Kölliker's organ. Innervation of the developing organ of Corti was disrupted with no obvious spiral bundles. The ectopic patches were also innervated. All the extra hair cells (HCs) within the sensory epithelium and Kölliker's organ contained mechanoelectrical transduction channels as indicated by rapid uptake of FM1-43. The excessive numbers of HCs were still present in the adult Ebf1 conditional knockout (cKO) animal. The animals had no detectable auditory brainstem response (ABR) suggesting that this gene is essential for hearing development.

20.
Development ; 2024 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-39101673

RESUMEN

The dorsal aorta (DA) is the first major blood vessel to develop in the embryonic cardiovascular system. Its formation is governed by a coordinated process involving the migration, specification, and arrangement of angioblasts into arterial and venous lineages, a process conserved across species. While vascular endothelial growth factor a (VEGF-A) drives DA specification and formation, the kinases involved in this process remain ambiguous. Thus, we investigated the role of protein kinase B, Akt, in zebrafish by generating a quadruple mutant (aktΔ/Δ), where expression and activity of all akt genes-akt 1, 2, 3a, and 3b are strongly decreased. Live imaging of developing aktΔ/Δ DA uncovers early arteriovenous malformations. Single-cell RNA sequencing analysis of aktΔ/Δ endothelial cells corroborates the impairment of arterial, yet not venous, cell specification. Notably, endothelial specific expression of ligand-independent activation of Notch or constitutively active Akt1 were sufficient to reestablish normal arterial specification in aktΔ/Δ. The Akt-loss-of-function mutant unveils that Akt kinase can act upstream of Notch in arterial endothelial cells, and is involved in proper embryonic artery specification. This sheds light on cardiovascular development, revealing a mechanism behind congenital malformations.

SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda