Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 86
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Annu Rev Genomics Hum Genet ; 24: 203-223, 2023 08 25.
Artículo en Inglés | MEDLINE | ID: mdl-37624665

RESUMEN

While the neural crest cell population gives rise to an extraordinary array of derivatives, including elements of the craniofacial skeleton, skin pigmentation, and peripheral nervous system, it is today increasingly recognized that Schwann cell precursors are also multipotent. Two mammalian paralogs of the SWI/SNF (switch/sucrose nonfermentable) chromatin-remodeling complexes, BAF (Brg1-associated factors) and PBAF (polybromo-associated BAF), are critical for neural crest specification during normal mammalian development. There is increasing evidence that pathogenic variants in components of the BAF and PBAF complexes play central roles in the pathogenesis of neural crest-derived tumors. Transgenic mouse models demonstrate a temporal window early in development where pathogenic variants in Smarcb1 result in the formation of aggressive, poorly differentiated tumors, such as rhabdoid tumors. By contrast, later in development, homozygous inactivation of Smarcb1 requires additional pathogenic variants in tumor suppressor genes to drive the development of differentiated adult neoplasms derived from the neural crest, which have a comparatively good prognosis in humans.


Asunto(s)
Agresión , Cresta Neural , Adulto , Animales , Ratones , Humanos , Diferenciación Celular/genética , Homocigoto , Ratones Transgénicos , Mamíferos
2.
Semin Cell Dev Biol ; 138: 1-14, 2023 03 30.
Artículo en Inglés | MEDLINE | ID: mdl-35941042

RESUMEN

The neural crest (NC) is an emblematic population of embryonic stem-like cells with remarkable migratory ability. These distinctive attributes have inspired the curiosity of developmental biologists for over 150 years, however only recently the regulatory mechanisms controlling the complex features of the NC have started to become elucidated at genomic scales. Regulatory control of NC development is achieved through combinatorial transcription factor binding and recruitment of associated transcriptional complexes to distal cis-regulatory elements. Together, they regulate when, where and to what extent transcriptional programmes are actively deployed, ultimately shaping ontogenetic processes. Here, we discuss how transcriptional networks control NC ontogeny, with a special emphasis on the molecular mechanisms underlying specification of the cephalic NC. We also cover emerging properties of transcriptional regulation revealed in diverse developmental systems, such as the role of three-dimensional conformation of chromatin, and how they are involved in the regulation of NC ontogeny. Finally, we highlight how advances in deciphering the NC transcriptional network have afforded new insights into the molecular basis of human diseases.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Cresta Neural , Humanos , Cresta Neural/metabolismo , Redes Reguladoras de Genes , Neurogénesis , Células Madre Embrionarias
3.
Genes Dev ; 31(1): 18-33, 2017 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-28096186

RESUMEN

The intratumor microenvironment generates phenotypically distinct but interconvertible malignant cell subpopulations that fuel metastatic spread and therapeutic resistance. Whether different microenvironmental cues impose invasive or therapy-resistant phenotypes via a common mechanism is unknown. In melanoma, low expression of the lineage survival oncogene microphthalmia-associated transcription factor (MITF) correlates with invasion, senescence, and drug resistance. However, how MITF is suppressed in vivo and how MITF-low cells in tumors escape senescence are poorly understood. Here we show that microenvironmental cues, including inflammation-mediated resistance to adoptive T-cell immunotherapy, transcriptionally repress MITF via ATF4 in response to inhibition of translation initiation factor eIF2B. ATF4, a key transcription mediator of the integrated stress response, also activates AXL and suppresses senescence to impose the MITF-low/AXL-high drug-resistant phenotype observed in human tumors. However, unexpectedly, without translation reprogramming an ATF4-high/MITF-low state is insufficient to drive invasion. Importantly, translation reprogramming dramatically enhances tumorigenesis and is linked to a previously unexplained gene expression program associated with anti-PD-1 immunotherapy resistance. Since we show that inhibition of eIF2B also drives neural crest migration and yeast invasiveness, our results suggest that translation reprogramming, an evolutionarily conserved starvation response, has been hijacked by microenvironmental stress signals in melanoma to drive phenotypic plasticity and invasion and determine therapeutic outcome.


Asunto(s)
Plasticidad de la Célula/genética , Reprogramación Celular/genética , Resistencia a Antineoplásicos/genética , Regulación Neoplásica de la Expresión Génica/genética , Melanoma/genética , Factor de Transcripción Asociado a Microftalmía/genética , Biosíntesis de Proteínas/genética , Animales , Microambiente Celular , Evolución Molecular , Retroalimentación Fisiológica , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Glutamina/farmacología , Humanos , Inmunoterapia , Melanoma/tratamiento farmacológico , Melanoma/metabolismo , Invasividad Neoplásica/genética , Cresta Neural/citología , Fenotipo , Factores de Transcripción/metabolismo , Pez Cebra/embriología
4.
Annu Rev Cell Dev Biol ; 26: 581-603, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-19575671

RESUMEN

The neural crest is a multipotent stem cell–like population that gives rise to a wide range of derivatives in the vertebrate embryo including elements of the craniofacial skeleton and peripheral nervous system as well as melanocytes. The neural crest forms in a series of regulatory steps that include induction and specification of the prospective neural crest territory–neural plate border, specification of bona fide neural crest progenitors, and differentiation into diverse derivatives. These individual processes during neural crest ontogeny are controlled by regulatory circuits that can be assembled into a hierarchical gene regulatory network (GRN). Here we present an overview of the GRN that orchestrates the formation of cranial neural crest cells. Formulation of this network relies on information largely inferred from gene perturbation studies performed in several vertebrate model organisms. Our representation of the cranial neural crest GRN also includes information about direct regulatory interactions obtained from the cis-regulatory analyses performed to date, which increases the resolution of the architectural circuitry within the network.


Asunto(s)
Redes Reguladoras de Genes , Cresta Neural/metabolismo , Animales , Movimiento Celular , Regulación del Desarrollo de la Expresión Génica , Cresta Neural/citología , Vertebrados/embriología
5.
Blood ; 136(3): 269-278, 2020 07 16.
Artículo en Inglés | MEDLINE | ID: mdl-32396940

RESUMEN

The oxygen transport function of hemoglobin (HB) is thought to have arisen ∼500 million years ago, roughly coinciding with the divergence between jawless (Agnatha) and jawed (Gnathostomata) vertebrates. Intriguingly, extant HBs of jawless and jawed vertebrates were shown to have evolved twice, and independently, from different ancestral globin proteins. This raises the question of whether erythroid-specific expression of HB also evolved twice independently. In all jawed vertebrates studied to date, one of the HB gene clusters is linked to the widely expressed NPRL3 gene. Here we show that the nprl3-linked hb locus of a jawless vertebrate, the river lamprey (Lampetra fluviatilis), shares a range of structural and functional properties with the equivalent jawed vertebrate HB locus. Functional analysis demonstrates that an erythroid-specific enhancer is located in intron 7 of lamprey nprl3, which corresponds to the NPRL3 intron 7 MCS-R1 enhancer of jawed vertebrates. Collectively, our findings signify the presence of an nprl3-linked multiglobin gene locus, which contains a remote enhancer that drives globin expression in erythroid cells, before the divergence of jawless and jawed vertebrates. Different globin genes from this ancestral cluster evolved in the current NPRL3-linked HB genes in jawless and jawed vertebrates. This provides an explanation of the enigma of how, in different species, globin genes linked to the same adjacent gene could undergo convergent evolution.


Asunto(s)
Eritrocitos/metabolismo , Evolución Molecular , Proteínas de Peces , Regulación de la Expresión Génica/fisiología , Hemoglobinas , Lampreas , Animales , Proteínas de Peces/biosíntesis , Proteínas de Peces/genética , Hemoglobinas/biosíntesis , Hemoglobinas/genética , Lampreas/genética , Lampreas/metabolismo , Familia de Multigenes
6.
Development ; 145(4)2018 02 23.
Artículo en Inglés | MEDLINE | ID: mdl-29386245

RESUMEN

CRISPR/Cas9 genome engineering has revolutionised all aspects of biological research, with epigenome engineering transforming gene regulation studies. Here, we present an optimised, adaptable toolkit enabling genome and epigenome engineering in the chicken embryo, and demonstrate its utility by probing gene regulatory interactions mediated by neural crest enhancers. First, we optimise novel efficient guide-RNA mini expression vectors utilising chick U6 promoters, provide a strategy for rapid somatic gene knockout and establish a protocol for evaluation of mutational penetrance by targeted next-generation sequencing. We show that CRISPR/Cas9-mediated disruption of transcription factors causes a reduction in their cognate enhancer-driven reporter activity. Next, we assess endogenous enhancer function using both enhancer deletion and nuclease-deficient Cas9 (dCas9) effector fusions to modulate enhancer chromatin landscape, thus providing the first report of epigenome engineering in a developing embryo. Finally, we use the synergistic activation mediator (SAM) system to activate an endogenous target promoter. The novel genome and epigenome engineering toolkit developed here enables manipulation of endogenous gene expression and enhancer activity in chicken embryos, facilitating high-resolution analysis of gene regulatory interactions in vivo.


Asunto(s)
Sistemas CRISPR-Cas/genética , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas/genética , Epigenómica/métodos , Ingeniería Genética/métodos , Animales , Embrión de Pollo , Pollos/genética , Clonación de Organismos , Electroporación , Técnica del Anticuerpo Fluorescente , Expresión Génica , Hibridación in Situ , Reacción en Cadena de la Polimerasa
7.
J Biol Chem ; 294(34): 12599-12609, 2019 08 23.
Artículo en Inglés | MEDLINE | ID: mdl-31270209

RESUMEN

Cholesterol constitutes ∼30-40% of the mammalian plasma membrane, a larger fraction than of any other single component. It is a major player in numerous signaling processes as well as in shaping molecular membrane architecture. However, our knowledge of the dynamics of cholesterol in the plasma membrane is limited, restricting our understanding of the mechanisms regulating its involvement in cell signaling. Here, we applied advanced fluorescence imaging and spectroscopy approaches on in vitro (model membranes) and in vivo (live cells and embryos) membranes as well as in silico analysis to systematically study the nanoscale dynamics of cholesterol in biological membranes. Our results indicate that cholesterol diffuses faster than phospholipids in live membranes, but not in model membranes. Interestingly, a detailed statistical diffusion analysis suggested two-component diffusion for cholesterol in the plasma membrane of live cells. One of these components was similar to a freely diffusing phospholipid analogue, whereas the other one was significantly faster. When a cholesterol analogue was localized to the outer leaflet only, the fast diffusion of cholesterol disappeared, and it diffused similarly to phospholipids. Overall, our results suggest that cholesterol diffusion in the cell membrane is heterogeneous and that this diffusional heterogeneity is due to cholesterol's nanoscale interactions and localization in the membrane.


Asunto(s)
Membrana Celular/química , Colesterol/análisis , Simulación de Dinámica Molecular , Nanotecnología , Animales , Células CHO , Membrana Celular/metabolismo , Células Cultivadas , Colesterol/metabolismo , Cricetulus , Difusión , Femenino , Masculino , Método de Montecarlo , Espectrometría de Fluorescencia , Pez Cebra
8.
Glia ; 68(12): 2550-2584, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32857879

RESUMEN

Olfactory ensheathing cells (OECs) are neural crest-derived glia that ensheath bundles of olfactory axons from their peripheral origins in the olfactory epithelium to their central targets in the olfactory bulb. We took an unbiased laser microdissection and differential RNA-seq approach, validated by in situ hybridization, to identify candidate molecular mechanisms underlying mouse OEC development and differences with the neural crest-derived Schwann cells developing on other peripheral nerves. We identified 25 novel markers for developing OECs in the olfactory mucosa and/or the olfactory nerve layer surrounding the olfactory bulb, of which 15 were OEC-specific (that is, not expressed by Schwann cells). One pan-OEC-specific gene, Ptprz1, encodes a receptor-like tyrosine phosphatase that blocks oligodendrocyte differentiation. Mutant analysis suggests Ptprz1 may also act as a brake on OEC differentiation, and that its loss disrupts olfactory axon targeting. Overall, our results provide new insights into OEC development and the diversification of neural crest-derived glia.


Asunto(s)
Microdisección , Transcriptoma , Animales , Diferenciación Celular , Células Cultivadas , Rayos Láser , Ratones , Neuroglía , Bulbo Olfatorio , Mucosa Olfatoria
9.
Nat Rev Mol Cell Biol ; 9(7): 557-68, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18523435

RESUMEN

The neural crest is a multipotent, migratory cell population that is unique to vertebrate embryos and gives rise to many derivatives, ranging from the peripheral nervous system to the craniofacial skeleton and pigment cells. A multimodule gene regulatory network mediates the complex process of neural crest formation, which involves the early induction and maintenance of the precursor pool, emigration of the neural crest progenitors from the neural tube via an epithelial to mesenchymal transition, migration of progenitor cells along distinct pathways and overt differentiation into diverse cell types. Here, we review our current understanding of these processes and discuss the molecular players that are involved in the neural crest gene regulatory network.


Asunto(s)
Diferenciación Celular/fisiología , Redes Reguladoras de Genes , Cresta Neural/embriología , Animales , Ciclo Celular/fisiología , Movimiento Celular/fisiología , Inducción Embrionaria , Uniones Comunicantes/metabolismo , Regulación del Desarrollo de la Expresión Génica , Morfogénesis , Cresta Neural/anatomía & histología , Cresta Neural/fisiología , Transducción de Señal/fisiología , Células Madre/citología , Células Madre/fisiología , Factores de Transcripción/metabolismo
11.
Genes Dev ; 26(21): 2380-5, 2012 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-23124063

RESUMEN

Here, we explore whether silencing via promoter DNA methylation plays a role in neural versus neural crest cell lineage decisions. We show that DNA methyltransferase3A (DNMT3A) promotes neural crest specification by directly mediating repression of neural genes like Sox2 and Sox3. DNMT3A is expressed in the neural plate border, and its knockdown causes ectopic Sox2 and Sox3 expression at the expense of neural crest markers. In vivo chromatin immunoprecipitation of neural folds demonstrates that DNMT3A specifically associates with CpG islands in the Sox2 and Sox3 promoter regions, resulting in their repression by methylation. Thus, DNMT3A functions as a molecular switch, repressing neural to favor neural crest cell fate.


Asunto(s)
Diferenciación Celular , ADN (Citosina-5-)-Metiltransferasas/genética , ADN (Citosina-5-)-Metiltransferasas/metabolismo , Regulación del Desarrollo de la Expresión Génica , Cresta Neural/citología , Tubo Neural/citología , Animales , Linaje de la Célula , Embrión de Pollo , Islas de CpG/genética , Metilación de ADN , ADN Metiltransferasa 3A , Técnicas de Silenciamiento del Gen , Silenciador del Gen , Cresta Neural/enzimología , Tubo Neural/enzimología
12.
Development ; 143(19): 3632-3637, 2016 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-27702788

RESUMEN

In situ hybridization methods are used across the biological sciences to map mRNA expression within intact specimens. Multiplexed experiments, in which multiple target mRNAs are mapped in a single sample, are essential for studying regulatory interactions, but remain cumbersome in most model organisms. Programmable in situ amplifiers based on the mechanism of hybridization chain reaction (HCR) overcome this longstanding challenge by operating independently within a sample, enabling multiplexed experiments to be performed with an experimental timeline independent of the number of target mRNAs. To assist biologists working across a broad spectrum of organisms, we demonstrate multiplexed in situ HCR in diverse imaging settings: bacteria, whole-mount nematode larvae, whole-mount fruit fly embryos, whole-mount sea urchin embryos, whole-mount zebrafish larvae, whole-mount chicken embryos, whole-mount mouse embryos and formalin-fixed paraffin-embedded human tissue sections. In addition to straightforward multiplexing, in situ HCR enables deep sample penetration, high contrast and subcellular resolution, providing an incisive tool for the study of interlaced and overlapping expression patterns, with implications for research communities across the biological sciences.


Asunto(s)
Hibridación in Situ/métodos , ARN Mensajero/metabolismo , Animales , Drosophila , Embrión no Mamífero/metabolismo , Humanos , Pez Cebra
13.
Methods ; 150: 24-31, 2018 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-30076893

RESUMEN

Interrogation of gene regulatory circuits in complex organisms requires precise and robust methods to label cell-types for profiling of target proteins in a tissue-specific fashion as well as data analysis to understand interconnections within the circuits. There are several strategies for obtaining cell-type and subcellular specific genome-wide data. We have developed a methodology, termed "biotagging" that uses tissue-specific, genetically encoded components to biotinylate target proteins, enabling in depth genome-wide profiling in zebrafish. We have refined protocols to use the biotagging approach that led to enhanced isolation of coding and non-coding RNAs from ribosomes and nuclei of genetically defined cell-types. The ability to study both the actively translated and transcribed transcriptome in the same cell population, coupled to genomic accessibility assays has enabled the study of cell-type specific gene regulatory circuits in zebrafish due to the high signal-to-noise achieved via its stringent purification protocol. Here, we provide detailed methods to isolate, profile and analyze cell-type specific polyribosome and nuclear transcriptome in zebrafish.


Asunto(s)
Biotinilación/métodos , Perfilación de la Expresión Génica/métodos , Coloración y Etiquetado/métodos , Pez Cebra/genética , Animales , Fraccionamiento Celular , Redes Reguladoras de Genes/genética , Polirribosomas/genética , Polirribosomas/metabolismo , ARN/aislamiento & purificación , ARN/metabolismo , Transcriptoma/genética , Pez Cebra/metabolismo
14.
PLoS Biol ; 13(2): e1002051, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25665164

RESUMEN

As some of the most widely utilised intercellular signalling molecules, transforming growth factor ß (TGFß) superfamily members play critical roles in normal development and become disrupted in human disease. Establishing appropriate levels of TGFß signalling involves positive and negative feedback, which are coupled and driven by the same signal transduction components (R-Smad transcription factor complexes), but whether and how the regulation of the two can be distinguished are unknown. Genome-wide comparison of published ChIP-seq datasets suggests that LIM domain binding proteins (Ldbs) co-localise with R-Smads at a substantial subset of R-Smad target genes including the locus of inhibitory Smad7 (I-Smad7), which mediates negative feedback for TGFß signalling. We present evidence suggesting that zebrafish Ldb2a binds and directly activates the I-Smad7 gene, whereas it binds and represses the ligand gene, Squint (Sqt), which drives positive feedback. Thus, the fine tuning of TGFß signalling derives from positive and negative control by Ldb2a. Expression of ldb2a is itself activated by TGFß signals, suggesting potential feed-forward loops that might delay the negative input of Ldb2a to the positive feedback, as well as the positive input of Ldb2a to the negative feedback. In this way, precise gene expression control by Ldb2a enables an initial build-up of signalling via a fully active positive feedback in the absence of buffering by the negative feedback. In Ldb2a-deficient zebrafish embryos, homeostasis of TGFß signalling is perturbed and signalling is stably enhanced, giving rise to excess mesoderm and endoderm, an effect that can be rescued by reducing signalling by the TGFß family members, Nodal and BMP. Thus, Ldb2a is critical to the homeostatic control of TGFß signalling and thereby embryonic patterning.


Asunto(s)
Tipificación del Cuerpo/genética , Retroalimentación Fisiológica , Proteínas con Dominio LIM/genética , Ligandos de Señalización Nodal/metabolismo , Proteína smad7/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo , Animales , Secuencia de Bases , Embrión no Mamífero , Endodermo/citología , Endodermo/embriología , Endodermo/metabolismo , Regulación del Desarrollo de la Expresión Génica , Humanos , Proteínas con Dominio LIM/antagonistas & inhibidores , Proteínas con Dominio LIM/deficiencia , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Mesodermo/citología , Mesodermo/embriología , Mesodermo/metabolismo , Ratones , Microinyecciones , Datos de Secuencia Molecular , Morfolinos/genética , Morfolinos/metabolismo , Ligandos de Señalización Nodal/genética , Alineación de Secuencia , Transducción de Señal , Proteína smad7/genética , Transcripción Genética , Factor de Crecimiento Transformador beta/genética , Pez Cebra , Proteínas de Pez Cebra/antagonistas & inhibidores , Proteínas de Pez Cebra/deficiencia
15.
Genome Res ; 24(2): 281-90, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24389048

RESUMEN

The neural crest is an embryonic stem cell population that gives rise to a multitude of derivatives. In particular, the cranial neural crest (CNC) is unique in its ability to contribute to both facial skeleton and peripheral ganglia. To gain further insight into the molecular underpinnings that distinguish the CNC from other embryonic tissues, we have utilized a CNC-specific enhancer as a tool to isolate a pure, region-specific NC subpopulation for transcriptional profiling. The resulting data set reveals previously unknown transcription factors and signaling pathways that may influence the CNC's ability to migrate and/or differentiate into unique derivatives. To elaborate on the CNC gene regulatory network, we evaluated the effects of knocking down known neural plate border genes and early neural crest specifier genes on selected neural crest-enriched transcripts. The results suggest that ETS1 and SOX9 may act as pan-neural crest regulators of the migratory CNC. Taken together, our analysis provides unprecedented characterization of the migratory CNC transcriptome and identifies new links in the gene regulatory network responsible for development of this critical cell population.


Asunto(s)
Redes Reguladoras de Genes , Proteína Proto-Oncogénica c-ets-1/genética , Factor de Transcripción SOX9/genética , Cráneo/crecimiento & desarrollo , Animales , Embrión de Pollo , Células Madre Embrionarias , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Genoma , Cresta Neural/crecimiento & desarrollo , Proteína Proto-Oncogénica c-ets-1/metabolismo , Factor de Transcripción SOX9/metabolismo , Transducción de Señal/genética
16.
Dev Biol ; 397(2): 282-92, 2015 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-25286121

RESUMEN

Members of the Sox family of transcription factors play a variety of critical developmental roles in both vertebrates and invertebrates. Whereas SoxBs and SoxEs are involved in neural and neural crest development, respectively, far less is known about members of the SoxC subfamily. To address this from an evolutionary perspective, we compare expression and function of SoxC genes in neural crest cells and their derivatives in lamprey (Petromyzon marinus), a basal vertebrate, to frog (Xenopus laevis). Analysis of transcript distribution reveals conservation of lamprey and X. laevis SoxC expression in premigratory neural crest, branchial arches, and cranial ganglia. Moreover, morpholino-mediated loss-of-function of selected SoxC family members demonstrates essential roles in aspects of neural crest development in both organisms. The results suggest important and conserved functions of SoxC genes during vertebrate evolution and a particularly critical, previously unrecognized role in early neural crest specification.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica/fisiología , Cresta Neural/embriología , Placa Neural/embriología , Petromyzon/embriología , Factores de Transcripción SOXC/metabolismo , Xenopus laevis/embriología , Animales , Clonación Molecular , Cartilla de ADN/genética , ADN Complementario/genética , Técnicas de Silenciamiento del Gen , Hibridación in Situ , Cresta Neural/metabolismo , Placa Neural/metabolismo , Oligonucleótidos/genética , Filogenia , beta-Galactosidasa
17.
Dev Biol ; 385(2): 405-16, 2014 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-24513489

RESUMEN

Cranial neurogenic placodes and the neural crest make essential contributions to key adult characteristics of all vertebrates, including the paired peripheral sense organs and craniofacial skeleton. Neurogenic placode development has been extensively characterized in representative jawed vertebrates (gnathostomes) but not in jawless fishes (agnathans). Here, we use in vivo lineage tracing with DiI, together with neuronal differentiation markers, to establish the first detailed fate-map for placode-derived sensory neurons in a jawless fish, the sea lamprey Petromyzon marinus, and to confirm that neural crest cells in the lamprey contribute to the cranial sensory ganglia. We also show that a pan-Pax3/7 antibody labels ophthalmic trigeminal (opV, profundal) placode-derived but not maxillomandibular trigeminal (mmV) placode-derived neurons, mirroring the expression of gnathostome Pax3 and suggesting that Pax3 (and its single Pax3/7 lamprey ortholog) is a pan-vertebrate marker for opV placode-derived neurons. Unexpectedly, however, our data reveal that mmV neuron precursors are located in two separate domains at neurula stages, with opV neuron precursors sandwiched between them. The different branches of the mmV nerve are not comparable between lampreys and gnatho-stomes, and spatial segregation of mmV neuron precursor territories may be a derived feature of lampreys. Nevertheless, maxillary and mandibular neurons are spatially segregated within gnathostome mmV ganglia, suggesting that a more detailed investigation of gnathostome mmV placode development would be worthwhile. Overall, however, our results highlight the conservation of cranial peripheral sensory nervous system development across vertebrates, yielding insight into ancestral vertebrate traits.


Asunto(s)
Ganglios Sensoriales/embriología , Petromyzon/embriología , Animales , Linaje de la Célula , Ganglios Sensoriales/citología , Cresta Neural/embriología , Neuronas/citología , Factores de Transcripción Paired Box/inmunología , Cráneo
18.
PLoS Genet ; 8(12): e1003142, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23284303

RESUMEN

The critical stem cell transcription factor FoxD3 is expressed by the premigratory and migrating neural crest, an embryonic stem cell population that forms diverse derivatives. Despite its important role in development and stem cell biology, little is known about what mediates FoxD3 activity in these cells. We have uncovered two FoxD3 enhancers, NC1 and NC2, that drive reporter expression in spatially and temporally distinct manners. Whereas NC1 activity recapitulates initial FoxD3 expression in the cranial neural crest, NC2 activity recapitulates initial FoxD3 expression at vagal/trunk levels while appearing only later in migrating cranial crest. Detailed mutational analysis, in vivo chromatin immunoprecipitation, and morpholino knock-downs reveal that transcription factors Pax7 and Msx1/2 cooperate with the neural crest specifier gene, Ets1, to bind to the cranial NC1 regulatory element. However, at vagal/trunk levels, they function together with the neural plate border gene, Zic1, which directly binds to the NC2 enhancer. These results reveal dynamic and differential regulation of FoxD3 in distinct neural crest subpopulations, suggesting that heterogeneity is encrypted at the regulatory level. Isolation of neural crest enhancers not only allows establishment of direct regulatory connections underlying neural crest formation, but also provides valuable tools for tissue specific manipulation and investigation of neural crest cell identity in amniotes.


Asunto(s)
Diferenciación Celular , Elementos de Facilitación Genéticos , Factores de Transcripción Forkhead , Cresta Neural , Proteínas de Pez Cebra , Pez Cebra , Animales , Movimiento Celular , Análisis Mutacional de ADN , Células Madre Embrionarias , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/metabolismo , Regulación del Desarrollo de la Expresión Génica , Genoma , Ratones , Cresta Neural/citología , Cresta Neural/metabolismo , Neuronas/citología , Neuronas/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Pez Cebra/embriología , Pez Cebra/genética , Pez Cebra/metabolismo , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo
19.
Development ; 138(17): 3689-98, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21775416

RESUMEN

The otic placode, a specialized region of ectoderm, gives rise to components of the inner ear and shares many characteristics with the neural crest, including expression of the key transcription factor Sox10. Here, we show that in avian embryos, a highly conserved cranial neural crest enhancer, Sox10E2, also controls the onset of Sox10 expression in the otic placode. Interestingly, we show that different combinations of paralogous transcription factors (Sox8, Pea3 and cMyb versus Sox9, Ets1 and cMyb) are required to mediate Sox10E2 activity in the ear and neural crest, respectively. Mutating their binding motifs within Sox10E2 greatly reduces enhancer activity in the ear. Moreover, simultaneous knockdown of Sox8, Pea3 and cMyb eliminates not only the enhancer-driven reporter expression, but also the onset of endogenous Sox10 expression in the ear. Rescue experiments confirm that the specific combination of Myb together with Sox8 and Pea3 is responsible for the onset of Sox10 expression in the otic placode, as opposed to Myb plus Sox9 and Ets1 for neural crest Sox10 expression. Whereas SUMOylation of Sox8 is not required for the initial onset of Sox10 expression, it is necessary for later otic vesicle formation. This new role of Sox8, Pea3 and cMyb in controlling Sox10 expression via a common otic/neural crest enhancer suggests an evolutionarily conserved function for the combination of paralogous transcription factors in these tissues of distinct embryological origin.


Asunto(s)
Elementos de Facilitación Genéticos/genética , Cresta Neural/metabolismo , Factores de Transcripción SOXE/genética , Animales , Embrión de Pollo , Pollos , Regulación del Desarrollo de la Expresión Génica/genética , Regulación del Desarrollo de la Expresión Génica/fisiología , Inmunohistoquímica , Hibridación in Situ , Sumoilación/genética , Sumoilación/fisiología
20.
Nature ; 453(7198): 1064-71, 2008 Jun 19.
Artículo en Inglés | MEDLINE | ID: mdl-18563158

RESUMEN

Lancelets ('amphioxus') are the modern survivors of an ancient chordate lineage, with a fossil record dating back to the Cambrian period. Here we describe the structure and gene content of the highly polymorphic approximately 520-megabase genome of the Florida lancelet Branchiostoma floridae, and analyse it in the context of chordate evolution. Whole-genome comparisons illuminate the murky relationships among the three chordate groups (tunicates, lancelets and vertebrates), and allow not only reconstruction of the gene complement of the last common chordate ancestor but also partial reconstruction of its genomic organization, as well as a description of two genome-wide duplications and subsequent reorganizations in the vertebrate lineage. These genome-scale events shaped the vertebrate genome and provided additional genetic variation for exploitation during vertebrate evolution.


Asunto(s)
Cordados/genética , Evolución Molecular , Genoma/genética , Animales , Cordados/clasificación , Secuencia Conservada , Elementos Transponibles de ADN/genética , Duplicación de Gen , Genes/genética , Ligamiento Genético , Humanos , Intrones/genética , Cariotipificación , Familia de Multigenes , Filogenia , Polimorfismo Genético/genética , Proteínas/genética , Sintenía , Factores de Tiempo , Vertebrados/clasificación , Vertebrados/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA