Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.209
Filtrar
1.
Neural Dev ; 19(1): 12, 2024 Jul 05.
Artigo em Inglês | MEDLINE | ID: mdl-38970093

RESUMO

BACKGROUND: A key step in nervous system development involves the coordinated control of neural progenitor specification and positioning. A long-standing model for the vertebrate CNS postulates that transient anatomical compartments - known as neuromeres - function to position neural progenitors along the embryonic anteroposterior neuraxis. Such neuromeres are apparent in the embryonic hindbrain - that contains six rhombomeres with morphologically apparent boundaries - but other neuromeres lack clear morphological boundaries and have instead been defined by different criteria, such as differences in gene expression patterns and the outcomes of transplantation experiments. Accordingly, the caudal hindbrain (CHB) posterior to rhombomere (r) 6 has been variably proposed to contain from two to five 'pseudo-rhombomeres', but the lack of comprehensive molecular data has precluded a detailed definition of such structures. METHODS: We used single-cell Multiome analysis, which allows simultaneous characterization of gene expression and chromatin state of individual cell nuclei, to identify and characterize CHB progenitors in the developing zebrafish CNS. RESULTS: We identified CHB progenitors as a transcriptionally distinct population, that also possesses a unique profile of accessible transcription factor binding motifs, relative to both r6 and the spinal cord. This CHB population can be subdivided along its dorsoventral axis based on molecular characteristics, but we do not find any molecular evidence that it contains multiple pseudo-rhombomeres. We further observe that the CHB is closely related to r6 at the earliest embryonic stages, but becomes more divergent over time, and that it is defined by a unique gene regulatory network. CONCLUSIONS: We conclude that the early CHB represents a single neuromere compartment that cannot be molecularly subdivided into pseudo-rhombomeres and that it may share an embryonic origin with r6.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Rombencéfalo , Peixe-Zebra , Animais , Peixe-Zebra/embriologia , Rombencéfalo/embriologia , Medula Espinal/embriologia , Análise de Célula Única , Neurogênese/fisiologia
2.
Cells ; 10(11)2021 10 20.
Artigo em Inglês | MEDLINE | ID: mdl-34831043

RESUMO

Studies have shown that the BH3-only domain Bad regulates brain development via the control of programmed cell death (PCD), but very few studies have addressed its effect on the molecular signaling of brain development in the system. In this work, we examined the novel role of zebrafish Bad in initial programmed cell death for brain morphogenesis through the priming of p53-mediated stress signaling. In a biological function study on the knockdown of Bad by morpholino oligonucleotides, at 24 h post-fertilization (hpf) Bad defects induced abnormal hindbrain development, as determined in a tissue section by means of HE staining which traced the damaged hindbrain. Then, genome-wide approaches for monitoring either the upregulation of apoptotic-related genes (11.8%) or the downregulation of brain development-related genes (29%) at the 24 hpf stage were implemented. The p53/caspase-8-mediated apoptotic death pathway was strongly involved, with the pathway being strongly reversed in a p53 mutant (p53M214K) line during Bad knockdown. Furthermore, we propose the involvement of a p53-mediated stress signal which is correlated with regulating Bad loss-mediated brain defects. We found that some major genes in brain development, such as crybb1, pva1b5, irx4a, pax7a, and fabp7a, were dramatically restored in the p53M214K line, and brain development recovered to return movement behavior to normal. Our findings suggest that Bad is required for (PCD) control, exerting a p53 stress signal on caspase-8/tBid-mediated death signaling and brain development-related gene regulation.


Assuntos
Apoptose/genética , Encéfalo/embriologia , Encéfalo/metabolismo , Transdução de Sinais , Proteína Supressora de Tumor p53/metabolismo , Proteínas de Peixe-Zebra/genética , Peixe-Zebra/embriologia , Peixe-Zebra/genética , Proteína de Morte Celular Associada a bcl/genética , Animais , Animais Geneticamente Modificados , Caspase 8/metabolismo , Regulação para Baixo/genética , Embrião não Mamífero/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Genoma , Mutação com Perda de Função/genética , Morfogênese/genética , Rombencéfalo/embriologia , Rombencéfalo/metabolismo , Natação , Proteínas de Peixe-Zebra/metabolismo , Proteína de Morte Celular Associada a bcl/metabolismo
3.
Dev Biol ; 479: 61-76, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34310923

RESUMO

Meis genes are known to play important roles in the hindbrain and neural crest cells of jawed vertebrates. To explore the roles of Meis genes in head development during evolution of vertebrates, we have identified four meis genes in the sea lamprey genome and characterized their patterns of expression and regulation, with a focus on the hindbrain and pharynx. Each of the lamprey meis genes displays temporally and spatially dynamic patterns of expression, some of which are coupled to rhombomeric domains in the developing hindbrain and select pharyngeal arches. Studies of Meis loci in mouse and zebrafish have identified enhancers that are bound by Hox and TALE (Meis and Pbx) proteins, implicating these factors in the direct regulation of Meis expression. We examined the lamprey meis loci and identified a series of cis-elements conserved between lamprey and jawed vertebrate meis genes. In transgenic reporter assays we demonstrated that these elements act as neural enhancers in lamprey embryos, directing reporter expression in appropriate domains when compared to expression of their associated endogenous meis gene. Sequence alignments reveal that these conserved elements are in similar relative positions of the meis loci and contain a series of consensus binding motifs for Hox and TALE proteins. This suggests that ancient Hox and TALE-responsive enhancers regulated expression of ancestral vertebrate meis genes in segmental domains in the hindbrain and have been retained in the meis loci during vertebrate evolution. The presence of conserved Meis, Pbx and Hox binding sites in these lamprey enhancers links Hox and TALE factors to regulation of lamprey meis genes in the developing hindbrain, indicating a deep ancestry for these regulatory interactions prior to the divergence of jawed and jawless vertebrates.


Assuntos
Lampreias/genética , Tubo Neural/embriologia , Rombencéfalo/embriologia , Animais , Sítios de Ligação , Padronização Corporal/genética , Sequência Conservada , Elementos Facilitadores Genéticos , Expressão Gênica/genética , Regulação da Expressão Gênica no Desenvolvimento/genética , Genes Homeobox/genética , Proteínas de Homeodomínio/metabolismo , Lampreias/metabolismo , Proteína Meis1/genética , Proteína Meis1/metabolismo , Crista Neural/metabolismo , Tubo Neural/metabolismo , Fator de Transcrição 1 de Leucemia de Células Pré-B/genética , Fator de Transcrição 1 de Leucemia de Células Pré-B/metabolismo , Rombencéfalo/metabolismo , Fatores de Transcrição/metabolismo
4.
J Comp Neurol ; 529(15): 3477-3496, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34180540

RESUMO

The avian auditory hindbrain is a longstanding model for studying neural circuit development. Information on gene regulatory network (GRN) components underlying this process, however, is scarce. Recently, the spatiotemporal expression of 12 microRNAs (miRNAs) was investigated in the mammalian auditory hindbrain. As a comparative study, we here investigated the spatiotemporal expression of the orthologous miRNAs during development of the chicken auditory hindbrain. All miRNAs were expressed both at E13, an immature stage, and P14, a mature stage of the auditory system. In most auditory nuclei, a homogeneous expression pattern was observed at both stages, like the mammalian system. An exception was the nucleus magnocellularis (NM). There, at E13, nine miRNAs showed a differential expression pattern along the cochleotopic axis with high expression at the rostromedial pole. One of them showed a gradient expression whereas eight showed a spatially selective expression at the rostral pole that reflected the different rhombomeric origins of this composite nucleus. The miRNA differential expression persisted in the NM to the mature stage, with the selective expression changed to linear gradients. Bioinformatics analysis predicted mRNA targets that are associated with neuronal developmental processes such as neurite and synapse organization, calcium and ephrin-Eph signaling, and neurotransmission. Overall, this first analysis of miRNAs in the chicken central auditory system reveals shared and strikingly distinct features between chicken and murine orthologues. The embryonic gradient expression of these GRN elements in the NM adds miRNA patterns to the list of cochleotopic and developmental gradients in the central auditory system.


Assuntos
Vias Auditivas/crescimento & desenvolvimento , Vias Auditivas/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , MicroRNAs/biossíntese , Rombencéfalo/crescimento & desenvolvimento , Rombencéfalo/metabolismo , Animais , Vias Auditivas/embriologia , Galinhas , Feminino , Masculino , MicroRNAs/genética , Rombencéfalo/embriologia
5.
PLoS Comput Biol ; 17(6): e1009077, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-34161317

RESUMO

The vertebrate hindbrain is segmented into rhombomeres (r) initially defined by distinct domains of gene expression. Previous studies have shown that noise-induced gene regulation and cell sorting are critical for the sharpening of rhombomere boundaries, which start out rough in the forming neural plate (NP) and sharpen over time. However, the mechanisms controlling simultaneous formation of multiple rhombomeres and accuracy in their sizes are unclear. We have developed a stochastic multiscale cell-based model that explicitly incorporates dynamic morphogenetic changes (i.e. convergent-extension of the NP), multiple morphogens, and gene regulatory networks to investigate the formation of rhombomeres and their corresponding boundaries in the zebrafish hindbrain. During pattern initiation, the short-range signal, fibroblast growth factor (FGF), works together with the longer-range morphogen, retinoic acid (RA), to specify all of these boundaries and maintain accurately sized segments with sharp boundaries. At later stages of patterning, we show a nonlinear change in the shape of rhombomeres with rapid left-right narrowing of the NP followed by slower dynamics. Rapid initial convergence improves boundary sharpness and segment size by regulating cell sorting and cell fate both independently and coordinately. Overall, multiple morphogens and tissue dynamics synergize to regulate the sizes and boundaries of multiple segments during development.


Assuntos
Padronização Corporal/fisiologia , Modelos Biológicos , Peixe-Zebra/embriologia , Animais , Padronização Corporal/genética , Biologia Computacional , Desenvolvimento Embrionário/genética , Desenvolvimento Embrionário/fisiologia , Fatores de Crescimento de Fibroblastos/fisiologia , Regulação da Expressão Gênica no Desenvolvimento , Substâncias de Crescimento/fisiologia , Rombencéfalo/citologia , Rombencéfalo/embriologia , Transdução de Sinais , Processos Estocásticos , Tretinoína/fisiologia , Peixe-Zebra/genética
6.
Ultrasound Obstet Gynecol ; 58(6): 864-874, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-33942916

RESUMO

OBJECTIVES: To describe the prenatal neuroimaging spectrum of rhombencephalosynapsis (RES) and criteria for its classification according to the severity of vermian anomaly. METHODS: In this multicenter retrospective study of fetuses with RES between 2002 and 2020, the medical records and brain ultrasound and magnetic resonance images were evaluated comprehensively to determine the severity of the vermian anomaly and the presence of associated brain findings. RES was classified, according to the pattern of vermian agenesis and the extent of the fusion of the hemispheres, as complete RES (complete absence of the vermis) or partial RES (further classified according to the part of the vermis that was missing and, consequently, the region of hemispheric fusion, as anterior, posterior, severe or mixed RES). Findings were compared between cases with complete and those with partial RES. RESULTS: Included in the study were 62 fetuses with a gestational age ranging between 12 and 37 weeks. Most had complete absence of the vermis (complete RES, 77.4% of cases), a 'round-shaped' cerebellum on axial views (72.6%) and a transverse cerebellar diameter (TCD) < 3rd centile (87.1%). Among the 22.6% of cases with partial RES, 6.5% were classified as severe partial, 6.5% as partial anterior, 8.1% as partial mixed and 1.6% as partial posterior. Half of these cases presented with normal or nearly normal cerebellar morphology and 28.5% had a TCD within the normal limits. Infratentorially, the fourth ventricle was abnormal in 88.7% of cases overall, and anomalies of the midbrain and pons were frequent (93.5% and 77.4%, respectively). Ventriculomegaly was observed in 80.6% of all cases, being more severe in cases with complete RES than in those with partial RES, with high rates of parenchymal and septal disruption. CONCLUSIONS: This study provides prenatal neuroimaging criteria for the diagnosis and classification of RES, and identification of related features, using ultrasound and magnetic resonance imaging. According to our findings, a diagnosis of RES should be considered in fetuses with a small TCD (severe cerebellar hypoplasia) and/or a round-shaped cerebellum on axial views, during the second or third trimester, especially when associated with ventriculomegaly. Partial RES is more common than previously thought, but presents an extreme diagnostic challenge, especially in cases with normal or nearly-normal cerebellar morphobiometric features. © 2021 International Society of Ultrasound in Obstetrics and Gynecology.


Assuntos
Anormalidades Múltiplas/diagnóstico por imagem , Vermis Cerebelar/anormalidades , Cerebelo/anormalidades , Anormalidades do Olho/diagnóstico por imagem , Doenças Renais Císticas/diagnóstico por imagem , Malformações do Sistema Nervoso/diagnóstico por imagem , Neuroimagem , Diagnóstico Pré-Natal/métodos , Retina/anormalidades , Rombencéfalo/anormalidades , Anormalidades Múltiplas/embriologia , Adulto , Vermis Cerebelar/diagnóstico por imagem , Vermis Cerebelar/embriologia , Cerebelo/diagnóstico por imagem , Cerebelo/embriologia , Anormalidades do Olho/embriologia , Feminino , Idade Gestacional , Humanos , Doenças Renais Císticas/embriologia , Imageamento por Ressonância Magnética , Imagem Multimodal , Malformações do Sistema Nervoso/embriologia , Gravidez , Retina/diagnóstico por imagem , Retina/embriologia , Estudos Retrospectivos , Rombencéfalo/diagnóstico por imagem , Rombencéfalo/embriologia , Índice de Gravidade de Doença , Ultrassonografia Pré-Natal
7.
Proc Natl Acad Sci U S A ; 118(19)2021 05 11.
Artigo em Inglês | MEDLINE | ID: mdl-33941697

RESUMO

Gap closure is a common morphogenetic process. In mammals, failure to close the embryonic hindbrain neuropore (HNP) gap causes fatal anencephaly. We observed that surface ectoderm cells surrounding the mouse HNP assemble high-tension actomyosin purse strings at their leading edge and establish the initial contacts across the embryonic midline. Fibronectin and laminin are present, and tensin 1 accumulates in focal adhesion-like puncta at this leading edge. The HNP gap closes asymmetrically, faster from its rostral than caudal end, while maintaining an elongated aspect ratio. Cell-based physical modeling identifies two closure mechanisms sufficient to account for tissue-level HNP closure dynamics: purse-string contraction and directional cell motion implemented through active crawling. Combining both closure mechanisms hastens gap closure and produces a constant rate of gap shortening. Purse-string contraction reduces, whereas crawling increases gap aspect ratio, and their combination maintains it. Closure rate asymmetry can be explained by asymmetric embryo tissue geometry, namely a narrower rostral gap apex, whereas biomechanical tension inferred from laser ablation is equivalent at the gaps' rostral and caudal closure points. At the cellular level, the physical model predicts rearrangements of cells at the HNP rostral and caudal extremes as the gap shortens. These behaviors are reproducibly live imaged in mouse embryos. Thus, mammalian embryos coordinate cellular- and tissue-level mechanics to achieve this critical gap closure event.


Assuntos
Embrião de Mamíferos/metabolismo , Crista Neural/metabolismo , Tubo Neural/metabolismo , Rombencéfalo/metabolismo , Anencefalia/embriologia , Anencefalia/genética , Anencefalia/metabolismo , Animais , Caderinas/metabolismo , Embrião de Mamíferos/embriologia , Feminino , Fibronectinas/metabolismo , Laminina/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Microscopia Confocal/métodos , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Crista Neural/embriologia , Tubo Neural/embriologia , Rombencéfalo/embriologia , Imagem com Lapso de Tempo/métodos
8.
Nat Genet ; 53(3): 379-391, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33603234

RESUMO

Rapid cellular responses to environmental stimuli are fundamental for development and maturation. Immediate early genes can be transcriptionally induced within minutes in response to a variety of signals. How their induction levels are regulated and their untimely activation by spurious signals prevented during development is poorly understood. We found that in developing sensory neurons, before perinatal sensory-activity-dependent induction, immediate early genes are embedded into a unique bipartite Polycomb chromatin signature, carrying active H3K27ac on promoters but repressive Ezh2-dependent H3K27me3 on gene bodies. This bipartite signature is widely present in developing cell types, including embryonic stem cells. Polycomb marking of gene bodies inhibits mRNA elongation, dampening productive transcription, while still allowing for fast stimulus-dependent mark removal and bipartite gene induction. We reveal a developmental epigenetic mechanism regulating the rapidity and amplitude of the transcriptional response to relevant stimuli, while preventing inappropriate activation of stimulus-response genes.


Assuntos
Cromatina/genética , Regulação da Expressão Gênica no Desenvolvimento , Genes Precoces , Proteínas do Grupo Polycomb/genética , Animais , Cromatina/metabolismo , Células-Tronco Embrionárias/fisiologia , Proteína Potenciadora do Homólogo 2 de Zeste/genética , Proteína Potenciadora do Homólogo 2 de Zeste/metabolismo , Epigênese Genética , Histonas/metabolismo , Camundongos Transgênicos , Mutação , Proteínas do Grupo Polycomb/metabolismo , Regiões Promotoras Genéticas , RNA Polimerase II/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Rombencéfalo/efeitos dos fármacos , Rombencéfalo/embriologia , Células Receptoras Sensoriais/fisiologia
9.
Nat Commun ; 12(1): 439, 2021 01 19.
Artigo em Inglês | MEDLINE | ID: mdl-33469032

RESUMO

Developmental genes are often regulated by multiple elements with overlapping activity. Yet, in most cases, the relative function of those elements and their contribution to endogenous gene expression remain poorly characterized. An example of this phenomenon is that distinct sets of enhancers have been proposed to direct Fgf8 in the limb apical ectodermal ridge and the midbrain-hindbrain boundary. Using in vivo CRISPR/Cas9 genome engineering, we functionally dissect this complex regulatory ensemble and demonstrate two distinct regulatory logics. In the apical ectodermal ridge, the control of Fgf8 expression appears distributed between different enhancers. In contrast, we find that in the midbrain-hindbrain boundary, one of the three active enhancers is essential while the other two are dispensable. We further dissect the essential midbrain-hindbrain boundary enhancer to reveal that it is also composed by a mixture of essential and dispensable modules. Cross-species transgenic analysis of this enhancer suggests that its composition may have changed in the vertebrate lineage.


Assuntos
Desenvolvimento Embrionário/genética , Elementos Facilitadores Genéticos/genética , Fator 8 de Crescimento de Fibroblasto/genética , Regulação da Expressão Gênica no Desenvolvimento , Engenharia Genética/métodos , Animais , Sistemas CRISPR-Cas/genética , Ectoderma/embriologia , Embrião de Mamíferos , Extremidades/embriologia , Estudos de Viabilidade , Feminino , Fator 8 de Crescimento de Fibroblasto/metabolismo , Redes Reguladoras de Genes , Masculino , Mesencéfalo/embriologia , Camundongos , Camundongos Transgênicos , Rombencéfalo/embriologia
11.
Development ; 147(22)2020 11 30.
Artigo em Inglês | MEDLINE | ID: mdl-33046507

RESUMO

The choroid plexus (ChP) is a secretory tissue that produces cerebrospinal fluid (CSF) secreted into the ventricular system. It is a monolayer of secretory, multiciliated epithelial cells derived from neuroepithelial progenitors and overlying a stroma of mesenchymal cells of mesodermal origin. Zfp423, which encodes a Kruppel-type zinc-finger transcription factor essential for cerebellar development and mutated in rare cases of cerebellar vermis hypoplasia/Joubert syndrome and other ciliopathies, is expressed in the hindbrain roof plate, from which the IV ventricle ChP arises, and, later, in mesenchymal cells, which give rise to the stroma and leptomeninges. Mouse Zfp423 mutants display a marked reduction of the hindbrain ChP (hChP), which: (1) fails to express established markers of its secretory function and genes implicated in its development and maintenance (Lmx1a and Otx2); (2) shows a perturbed expression of signaling pathways previously unexplored in hChP patterning (Wnt3); and (3) displays a lack of multiciliated epithelial cells and a profound dysregulation of master genes of multiciliogenesis (Gmnc). Our results propose that Zfp423 is a master gene and one of the earliest known determinants of hChP development.


Assuntos
Plexo Corióideo/embriologia , Proteínas de Ligação a DNA/metabolismo , Rombencéfalo/embriologia , Fatores de Transcrição/metabolismo , Animais , Plexo Corióideo/citologia , Proteínas de Ligação a DNA/genética , Proteínas com Homeodomínio LIM/genética , Proteínas com Homeodomínio LIM/metabolismo , Camundongos , Camundongos Mutantes , Fatores de Transcrição Otx/genética , Fatores de Transcrição Otx/metabolismo , Rombencéfalo/citologia , Fatores de Transcrição/genética , Proteína Wnt3/genética , Proteína Wnt3/metabolismo
12.
Circ Res ; 127(10): 1221-1232, 2020 10 23.
Artigo em Inglês | MEDLINE | ID: mdl-32791884

RESUMO

RATIONALE: Endothelial cells are thought to emerge de novo from the mesoderm to form the entire circulatory system. Recently, erythro-myeloid progenitors (EMPs) have been proposed to be another remarkable developmental origin for blood vessels in multiple organs, including the hindbrain, liver, lung, and heart, as demonstrated by lineage tracing studies using different genetic tools. These observations challenge the current consensus that intraembryonic vessels are thought to expand solely by the proliferation of preexisting endothelial cells. Resolution of this controversy over the developmental origin of endothelial cells is crucial for developing future therapeutics for vessel-dependent organ repair and regeneration. OBJECTIVE: To examine the contribution of EMPs to intraembryonic endothelial cells. METHODS AND RESULTS: We first used a transgenic mouse expressing a tamoxifen-inducible Mer-iCre fusion protein driven by the Csf1r (colony stimulating factor 1 receptor) promoter. Genetic lineage tracing based on Csf1r-Mer-iCre-Mer showed no contribution of EMPs to brain endothelial cells identified by several markers. We also generated a knock-in mouse line by inserting an internal ribosome entry site-iCre cassette into the 3' untranslated region of Csf1r gene to further investigate the cellular fates of EMPs. Similarly, we did not find any Csf1r-ires-iCre traced endothelial cells in brain, liver, lung, or heart in development either. Additionally, we found that Kit (KIT proto-oncogene receptor tyrosine kinase) was expressed not only in EMPs but also in embryonic hindbrain endothelial cells. Therefore, Kit promoter-driven recombinase, such as Kit-CreER, is a flawed tool for lineage tracing when examining the contribution of EMPs to hindbrain endothelial cells. We also traced CD45 (protein tyrosine phosphatase receptor type C; Ptprc)+ circulating EMPs and did not find any CD45 lineage-derived endothelial cells during development. CONCLUSIONS: Our study suggested that EMPs are not the origin of intraembryonic endothelial cells.


Assuntos
Linhagem da Célula , Células Endoteliais/citologia , Células Precursoras Eritroides/citologia , Animais , Endotélio Vascular/citologia , Endotélio Vascular/embriologia , Coração Fetal/citologia , Fígado/citologia , Fígado/embriologia , Pulmão/citologia , Pulmão/embriologia , Macrófagos/citologia , Mesoderma/citologia , Camundongos , Rombencéfalo/citologia , Rombencéfalo/embriologia
13.
Development ; 147(21)2020 08 25.
Artigo em Inglês | MEDLINE | ID: mdl-32747436

RESUMO

Fragile X mental retardation protein (FMRP) is an RNA-binding protein abundant in the nervous system. Functional loss of FMRP leads to sensory dysfunction and severe intellectual disabilities. In the auditory system, FMRP deficiency alters neuronal function and synaptic connectivity and results in perturbed processing of sound information. Nevertheless, roles of FMRP in embryonic development of the auditory hindbrain have not been identified. Here, we developed high-specificity approaches to genetically track and manipulate throughout development of the Atoh1+ neuronal cell type, which is highly conserved in vertebrates, in the cochlear nucleus of chicken embryos. We identified distinct FMRP-containing granules in the growing axons of Atoh1+ neurons and post-migrating NM cells. FMRP downregulation induced by CRISPR/Cas9 and shRNA techniques resulted in perturbed axonal pathfinding, delay in midline crossing, excess branching of neurites, and axonal targeting errors during the period of circuit development. Together, these results provide the first in vivo identification of FMRP localization and actions in developing axons of auditory neurons, and demonstrate the importance of investigating early embryonic alterations toward understanding the pathogenesis of neurodevelopmental disorders.


Assuntos
Vias Auditivas/embriologia , Vias Auditivas/metabolismo , Proteína do X Frágil da Deficiência Intelectual/metabolismo , Rombencéfalo/embriologia , Rombencéfalo/metabolismo , Animais , Axônios/metabolismo , Sequência de Bases , Sistemas CRISPR-Cas/genética , Embrião de Galinha , Galinhas , Dendritos/metabolismo , Células-Tronco Neurais/metabolismo , Terminações Pré-Sinápticas/metabolismo , RNA Interferente Pequeno/metabolismo , Sinapses/metabolismo , Fatores de Tempo
14.
Proc Natl Acad Sci U S A ; 117(32): 19544-19555, 2020 08 11.
Artigo em Inglês | MEDLINE | ID: mdl-32747566

RESUMO

Corresponding attributes of neural development and function suggest arthropod and vertebrate brains may have an evolutionarily conserved organization. However, the underlying mechanisms have remained elusive. Here, we identify a gene regulatory and character identity network defining the deutocerebral-tritocerebral boundary (DTB) in Drosophila This network comprises genes homologous to those directing midbrain-hindbrain boundary (MHB) formation in vertebrates and their closest chordate relatives. Genetic tracing reveals that the embryonic DTB gives rise to adult midbrain circuits that in flies control auditory and vestibular information processing and motor coordination, as do MHB-derived circuits in vertebrates. DTB-specific gene expression and function are directed by cis-regulatory elements of developmental control genes that include homologs of mammalian Zinc finger of the cerebellum and Purkinje cell protein 4Drosophila DTB-specific cis-regulatory elements correspond to regulatory sequences of human ENGRAILED-2, PAX-2, and DACHSHUND-1 that direct MHB-specific expression in the embryonic mouse brain. We show that cis-regulatory elements and the gene networks they regulate direct the formation and function of midbrain circuits for balance and motor coordination in insects and mammals. Regulatory mechanisms mediating the genetic specification of cephalic neural circuits in arthropods correspond to those in chordates, thereby implying their origin before the divergence of deuterostomes and ecdysozoans.


Assuntos
Evolução Molecular , Redes Reguladoras de Genes , Mesencéfalo/fisiologia , Animais , Comportamento Animal , Encéfalo/embriologia , Encéfalo/metabolismo , Encéfalo/fisiologia , Drosophila , Fator 8 de Crescimento de Fibroblasto/genética , Fator 8 de Crescimento de Fibroblasto/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Mesencéfalo/embriologia , Mesencéfalo/metabolismo , Camundongos , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Vias Neurais , Fatores de Transcrição Box Pareados/genética , Fatores de Transcrição Box Pareados/metabolismo , Sequências Reguladoras de Ácido Nucleico , Rombencéfalo/embriologia , Rombencéfalo/metabolismo , Rombencéfalo/fisiologia , Transdução de Sinais
15.
Semin Cell Dev Biol ; 107: 179-189, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32448645

RESUMO

Embryonic boundaries were first described in Drosophila, and then in vertebrate embryos, as cellular interfaces between compartments. They display signaling properties and in vertebrates might allocate cells fated to different anatomical structures, or cells that will play different functions over time. One of the vertebrate embryonic structures with boundaries is the hindbrain, the posterior brain vesicle, which is transitory segmented upon morphogenesis. The hindbrain is formed by iterative units called rhombomeres that constitute units of gene expression and cell-lineage compartments. Rhombomeric cells are segregated by interhombomeric boundaries, which are prefigured by sharp gene expression borders. Hindbrain boundaries were first described as static groups of cells. However, later discoveries demonstrated the dynamic behavior of this specific cell population. They play distinct functional properties during brain morphogenesis that partially overlap on time, starting as a mechanical barrier to prevent cell intermingling, becoming a signaling hub, to finally constitute a group of proliferating progenitors providing differentiated neurons to the system. In this review, I try to give a more functional overview of this segmentation process and in particular of hindbrain boundaries. I will discuss the new challenges in the field on how to integrate cell fate specification and morphogenesis during brain embryonic development.


Assuntos
Rombencéfalo/citologia , Rombencéfalo/embriologia , Animais , Proliferação de Células , Desenvolvimento Embrionário , Humanos , Mecanotransdução Celular , Modelos Biológicos , Filogenia
16.
Curr Top Dev Biol ; 139: 169-203, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32450960

RESUMO

In vertebrates, the hindbrain serves as a highly conserved complex coordination center for regulating many fundamental activities of the central nervous system, such as respiratory rhythms, sleep patterns and equilibrium, and it also plays an important role in craniofacial development. The basic ground plan that underlies the diverse functions of the hindbrain and its neural crest derivatives is established and patterned by a process of segmentation. Through a dynamic series of signaling and regulatory interactions the developing hindbrain is transiently compartmentalized into a set of seven segmental units, termed rhombomeres. The nested expression of the Hox family of transcription factors is tightly coupled to the process of segmentation and provides a molecular code for specifying the unique regional properties of the hindbrain and its neural crest derived craniofacial structures. The high degree of similarity in hindbrain architecture between diverse vertebrates has enabled cross-species regulatory analysis. This has facilitated the experimental assembly of the signaling and regulatory interactions, which underlie the process of segmentation, into a Hox-dependent gene regulatory network (GRN) model. This hindbrain GRN is a key regulatory feature of head patterning, conserved to the base of vertebrate evolution. This regulatory framework also serves as a basis for comparing and contrasting GRNs that govern cranial neural crest formation and axial patterning and provide insight into regulatory mechanisms associated with the evolution of novel vertebrate traits. The purpose of this review is to discuss the majorfeatures of the GRN for hindbrain segmentation and its relationship to the broader functional role of the hindbrain in patterning head development.


Assuntos
Padronização Corporal/genética , Regulação da Expressão Gênica no Desenvolvimento , Redes Reguladoras de Genes , Genes Homeobox/genética , Rombencéfalo/metabolismo , Animais , Evolução Molecular , Humanos , Crista Neural/embriologia , Crista Neural/metabolismo , Rombencéfalo/embriologia , Vertebrados/embriologia , Vertebrados/genética
17.
Biol Open ; 9(5)2020 05 07.
Artigo em Inglês | MEDLINE | ID: mdl-32295832

RESUMO

Human infantile-onset RNASET2-deficient cystic leukoencephalopathy is a Mendelian mimic of in utero cytomegalovirus brain infection with prenatally developing inflammatory brain lesions. We used an RNASET2-deficient zebrafish model to elucidate the underlying disease mechanisms. Mutant and wild-type zebrafish larvae brain development between 2 and 5 days post fertilization (dpf) was examined by confocal live imaging in fluorescent reporter lines of the major types of brain cells. In contrast to wild-type brains, RNASET2-deficient larvae displayed increased numbers of microglia with altered morphology, often containing inclusions of neurons. Furthermore, lysosomes within distinct populations of the myeloid cell lineage including microglia showed increased lysosomal staining. Neurons and oligodendrocyte precursor cells remained unaffected. This study provides a first look into the prenatal onset pathomechanisms of human RNASET2-deficient leukoencephalopathy, linking this inborn lysosomal disease to the innate immune system and other immune-related childhood encephalopathies like Aicardi-Goutières syndrome (AGS).


Assuntos
Estudos de Associação Genética , Predisposição Genética para Doença , Leucoencefalopatias/diagnóstico , Leucoencefalopatias/genética , Leucoencefalopatias/metabolismo , Microglia/metabolismo , Fenótipo , Ribonucleases/deficiência , Proteínas Supressoras de Tumor/deficiência , Animais , Apoptose , Suscetibilidade a Doenças , Estudos de Associação Genética/métodos , Humanos , Larva , Neurônios/metabolismo , Organogênese/genética , Rombencéfalo/anormalidades , Rombencéfalo/embriologia , Rombencéfalo/metabolismo , Peixe-Zebra
18.
Mayo Clin Proc ; 95(4): 738-746, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-32247347

RESUMO

OBJECTIVE: To assess our initial experience with prenatal restoration of hindbrain herniation following in utero repair of myelomeningocele (MMC). PATIENTS AND METHODS: Three consecutive patients with prenatally diagnosed MMC (between January 1, 2018 and September 30, 2018) were managed with open in utero surgery. As per institutional review board approval and following a protocol designed at the Mayo Clinic Maternal & Fetal Center, fetal intervention was offered between 19 0/7 and 25 6/7 weeks of gestation. Prenatal improvement of hindbrain herniation was the declared restorative end point. Obstetrical and perinatal outcomes were also assessed. RESULTS: Diagnosis of MMC was confirmed upon referral between 20 and 21 weeks' gestation by using fetal ultrasound and magnetic resonance imaging. In all cases reported here, the spinal defect was lumbosacral with evidence of hindbrain herniation. Open in utero MMC repair was performed between 24 and 25 weeks' gestation with no notable perioperative complications. Postprocedure fetal magnetic resonance imaging performed 6 weeks after in utero repair documented improvement of hindbrain herniation. Deliveries were at 37 weeks by cesarean section without complications. Most recent postnatal follow-ups were unremarkable at both 11 months (baby 1) and 3 months of age (baby 2), with mild ventriculomegaly. Antenatal and postnatal follow-up of baby 3 at 1 month of age was also unremarkable. CONCLUSION: Our study highlights the prenatal restoration of hindbrain herniation following in utero MMC repair in all cases presented here as an example of a prenatal regenerative therapy program in our institution.


Assuntos
Encefalocele/embriologia , Meningomielocele/embriologia , Medicina Regenerativa/métodos , Rombencéfalo/embriologia , Adulto , Encefalocele/cirurgia , Feminino , Feto/anormalidades , Feto/cirurgia , Humanos , Meningomielocele/cirurgia , Gravidez , Cuidado Pré-Natal/métodos , Medicina Regenerativa/classificação , Rombencéfalo/anormalidades , Rombencéfalo/cirurgia
19.
Development ; 147(6)2020 03 16.
Artigo em Inglês | MEDLINE | ID: mdl-32094115

RESUMO

Segmentation of the vertebrate hindbrain leads to the formation of rhombomeres, each with a distinct anteroposterior identity. Specialised boundary cells form at segment borders that act as a source or regulator of neuronal differentiation. In zebrafish, there is spatial patterning of neurogenesis in which non-neurogenic zones form at boundaries and segment centres, in part mediated by Fgf20 signalling. To further understand the control of neurogenesis, we have carried out single cell RNA sequencing of the zebrafish hindbrain at three different stages of patterning. Analyses of the data reveal known and novel markers of distinct hindbrain segments, of cell types along the dorsoventral axis, and of the transition of progenitors to neuronal differentiation. We find major shifts in the transcriptome of progenitors and of differentiating cells between the different stages analysed. Supervised clustering with markers of boundary cells and segment centres, together with RNA-seq analysis of Fgf-regulated genes, has revealed new candidate regulators of cell differentiation in the hindbrain. These data provide a valuable resource for functional investigations of the patterning of neurogenesis and the transition of progenitors to neuronal differentiation.


Assuntos
Padronização Corporal/genética , Rombencéfalo/embriologia , Rombencéfalo/metabolismo , Transcriptoma/fisiologia , Peixe-Zebra , Animais , Animais Geneticamente Modificados , Atlas como Assunto , Diferenciação Celular/genética , Embrião não Mamífero , Perfilação da Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento , Redes Reguladoras de Genes , Células-Tronco Neurais/citologia , Células-Tronco Neurais/metabolismo , Neurogênese/genética , Neurônios/citologia , Neurônios/fisiologia , Análise de Célula Única/métodos , Distribuição Tecidual , Peixe-Zebra/embriologia , Peixe-Zebra/genética , Peixe-Zebra/metabolismo
20.
Dev Dyn ; 249(3): 354-368, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31408233

RESUMO

BACKGROUND: The basic ground plan of vertebrate hindbrain is established through a process of segmentation, which generates eight transient lineage-restricted cellular compartments called rhombomeres (r). The segments adopt distinct individual identities in response to axial patterning signals. It is unclear whether signaling between rhombomeres plays a conserved role in regulating segmental patterning during hindbrain development. RESULTS: Using tissue manipulations of rhombomeres in chicken embryos, we have uncovered roles for r2 and r4 in regulating the expression of EphA4 in r3 and r5. Perturbations of signaling pathways reveal that these regulatory inputs from r2 and r4 into EphA4 expression are mediated independent of inputs from Krox20 through cues involving fibroblast growth factor (FGF) signaling. These interactions are stage dependent and are set up in embryos with <10 somites. CONCLUSIONS: We show that r2 and r4 function as temporally dynamic signaling centers in the early patterning of adjacent hindbrain segments and this activity is dependent upon the FGF pathway. These results reveal that inter-rhombomeric signaling is a conserved feature of the regulatory networks that control the specification of individual rhombomere identities in vertebrate hindbrain segmentation. However, the timing of when restricted domains of FGF signaling are coupled to formation of r4 may vary between the species.


Assuntos
Fatores de Crescimento de Fibroblastos/metabolismo , Receptor EphA4/metabolismo , Receptores de Fatores de Crescimento de Fibroblastos/metabolismo , Rombencéfalo/metabolismo , Animais , Embrião de Galinha , Fatores de Crescimento de Fibroblastos/genética , Hibridização In Situ , Receptor EphA4/genética , Receptores da Família Eph/genética , Receptores da Família Eph/metabolismo , Receptores de Fatores de Crescimento de Fibroblastos/genética , Rombencéfalo/embriologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...