Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 88
Filtrar
1.
Cell Rep Methods ; 1(4)2021 08 23.
Artigo em Inglês | MEDLINE | ID: mdl-34557863

RESUMO

Genetic loss and gain of function in mice have typically been studied by using knockout or knockin mice that take months to years to generate. To address this problem for the nervous system, we developed NEPTUNE (NEural Plate Targeting by in Utero NanoinjEction) to rapidly and flexibly transduce the neural plate with virus prior to neurulation, and thus manipulate the future nervous system. Stable integration in >95% of cells in the brain enabled long-term overexpression, and conditional expression was achieved by using cell-type-specific MiniPromoters. Knockdown of Olig2 by using NEPTUNE recapitulated the phenotype of Olig2 -/- embryos. We used NEPTUNE to investigate Sptbn2, mutations in which cause spinocerebellar ataxia type 5. Sptbn2 knockdown induced dose-dependent defects in the neural tube, embryonic turning, and abdominal wall closure, previously unreported functions for Sptbn2. NEPTUNE thus offers a rapid and cost-effective technique to test gene function in the nervous system and can reveal phenotypes incompatible with life.


Assuntos
Netuno , Defeitos do Tubo Neural , Camundongos , Animais , Tubo Neural/fisiologia , Encéfalo , Expressão Gênica
2.
Development ; 148(15)2021 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-34351410

RESUMO

The spinal cord receives input from peripheral sensory neurons and controls motor output by regulating muscle innervating motor neurons. These functions are carried out by neural circuits comprising molecularly distinct neuronal subtypes generated in a characteristic spatiotemporal arrangement from progenitors in the embryonic neural tube. To gain insight into the diversity and complexity of cells in the developing human neural tube, we used single-cell mRNA sequencing to profile cervical and thoracic regions in four human embryos of Carnegie stages (CS) CS12, CS14, CS17 and CS19 from gestational weeks 4-7. Analysis of progenitor and neuronal populations from the neural tube and dorsal root ganglia identified dozens of distinct cell types and facilitated the reconstruction of the differentiation pathways of specific neuronal subtypes. Comparison with mouse revealed overall similarity of mammalian neural tube development while highlighting some human-specific features. These data provide a catalogue of gene expression and cell type identity in the human neural tube that will support future studies of sensory and motor control systems. The data can be explored at https://shiny.crick.ac.uk/scviewer/neuraltube/.


Assuntos
Medula Espinal/fisiologia , Transcriptoma/genética , Transcriptoma/fisiologia , Animais , Diferenciação Celular/fisiologia , Embrião de Mamíferos/fisiologia , Gânglios Espinais/fisiologia , Expressão Gênica/genética , Perfilação da Expressão Gênica/métodos , Humanos , Camundongos , Neurônios Motores/fisiologia , Tubo Neural/fisiologia , Células Receptoras Sensoriais/fisiologia , Tórax/fisiologia
3.
Development ; 148(18)2021 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-34463728

RESUMO

The collective polarization of cellular structures and behaviors across a tissue plane is a near universal feature of epithelia known as planar cell polarity (PCP). This property is controlled by the core PCP pathway, which consists of highly conserved membrane-associated protein complexes that localize asymmetrically at cell junctions. Here, we introduce three new mouse models for investigating the localization and dynamics of transmembrane PCP proteins: Celsr1, Fz6 and Vangl2. Using the skin epidermis as a model, we characterize and verify the expression, localization and function of endogenously tagged Celsr1-3xGFP, Fz6-3xGFP and tdTomato-Vangl2 fusion proteins. Live imaging of Fz6-3xGFP in basal epidermal progenitors reveals that the polarity of the tissue is not fixed through time. Rather, asymmetry dynamically shifts during cell rearrangements and divisions, while global, average polarity of the tissue is preserved. We show using super-resolution STED imaging that Fz6-3xGFP and tdTomato-Vangl2 can be resolved, enabling us to observe their complex localization along junctions. We further explore PCP fusion protein localization in the trachea and neural tube, and discover new patterns of PCP expression and localization throughout the mouse embryo.


Assuntos
Polaridade Celular/fisiologia , Proteínas de Membrana/metabolismo , Animais , Padronização Corporal/fisiologia , Diagnóstico por Imagem/métodos , Embrião de Mamíferos/metabolismo , Embrião de Mamíferos/fisiologia , Células Epidérmicas/metabolismo , Células Epidérmicas/fisiologia , Epiderme/metabolismo , Epiderme/fisiologia , Epitélio/metabolismo , Epitélio/fisiologia , Receptores Frizzled/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Modelos Animais , Proteínas do Tecido Nervoso/metabolismo , Tubo Neural/metabolismo , Tubo Neural/fisiologia , Receptores Acoplados a Proteínas G/metabolismo , Transdução de Sinais/fisiologia , Traqueia/metabolismo , Traqueia/fisiologia
4.
Methods Mol Biol ; 2258: 193-203, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33340362

RESUMO

Neurally differentiating human pluripotent stem cells (hPSCs) possess the ability to self-organize into structures reminiscent of the developing fetal brain. In 2- and 3D cultures, this phenomenon initiates with formation of polarized areas of neural stem cells (NSCs), known as rosettes that resemble cross-sectional slices of the embryonic neural tube, i.e., the central nervous system (CNS) anlage. Thus, neural rosettes serve as an excellent starting point for bioengineering tissue models of all CNS tissues. Here, we provide detailed methods for bioengineering controlled induction of hPSC-derived neural assemblies with a biomimetic, singular neural rosette cytoarchitecture.


Assuntos
Células-Tronco Embrionárias Humanas/fisiologia , Células-Tronco Neurais/fisiologia , Tubo Neural/fisiologia , Neurogênese , Neurônios/fisiologia , Engenharia Tecidual , Materiais Biomiméticos , Técnicas de Cultura de Células , Linhagem Celular , Células-Tronco Embrionárias Humanas/metabolismo , Humanos , Imuno-Histoquímica , Microscopia , Morfogênese , Proteínas do Tecido Nervoso/metabolismo , Células-Tronco Neurais/metabolismo , Tubo Neural/citologia , Tubo Neural/metabolismo , Neurônios/metabolismo , Esferoides Celulares
5.
Dev Cell ; 52(3): 321-334.e6, 2020 02 10.
Artigo em Inglês | MEDLINE | ID: mdl-32049039

RESUMO

Epithelial fusion is a key process of morphogenesis by which tissue connectivity is established between adjacent epithelial sheets. A striking and poorly understood feature of this process is "zippering," whereby a fusion point moves directionally along an organ rudiment. Here, we uncover the molecular mechanism underlying zippering during mouse spinal neural tube closure. Fusion is initiated via local activation of integrin ß1 and focal anchorage of surface ectoderm cells to a shared point of fibronectin-rich basement membrane, where the neural folds first contact each other. Surface ectoderm cells undergo proximal junction shortening, establishing a transitory semi-rosette-like structure at the zippering point that promotes juxtaposition of cells across the midline enabling fusion propagation. Tissue-specific ablation of integrin ß1 abolishes the semi-rosette formation, preventing zippering and causing spina bifida. We propose integrin-mediated anchorage as an evolutionarily conserved mechanism of general relevance for zippering closure of epithelial gaps whose disturbance can produce clinically important birth defects.


Assuntos
Embrião de Mamíferos/fisiologia , Células Epiteliais/fisiologia , Adesões Focais , Integrina beta1/fisiologia , Crista Neural/embriologia , Tubo Neural/embriologia , Neurulação , Actomiosina/metabolismo , Animais , Fusão Celular , Embrião de Mamíferos/citologia , Embrião de Mamíferos/metabolismo , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Feminino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Morfogênese , Crista Neural/metabolismo , Crista Neural/fisiologia , Tubo Neural/metabolismo , Tubo Neural/fisiologia
6.
Med Hypotheses ; 129: 109264, 2019 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-31371090

RESUMO

Herein it is hypothesized that M2-like macrophages or pre-macrophages of fetal origin might play a central role in development and closure of the neural tube. Early in embryonic development, pre-macrophages arise from the fetal yolk sac and track through the bloodstream to reach diverse embryonic tissues, where they mature. Most of these macrophages exhibit an M2-like phenotype. The critical period for neural tube closure is contained within the period of yolk sac-derived pre-macrophage tracking and distribution, which poses a question: might these pre-macrophages or macrophages exert an influence on the closing neural tube? Evidence suggests that perturbations in macrophage polarization or M2 macrophage function might contribute to the failure of neural tube closure associated with diabetes mellitus, one carbon metabolism (including folic acid deficit), inositol, arachidonic acid, and sphingosine-1-phosphate, as well as in the teratogenicity of nitric acid, valproic acid, and fumonisin. The influence of each of these factors is interpreted in light of potential interactions with M2-like macrophages or macrophage progenitors on the developing neural tube. By placing these anti inflammatory macrophages at the center of various epigenetic, neurochemical, and signaling processes suspected to be involved in neural tube closure, potential associations are revealed between macrophages and embryonic structural developmental processes such as collagen and actin dynamics. The choice of this model is also an attempt to explain why some etiologies for failure of neural tube closure are rescued by folic acid, whereas other etiologies are rescued only by formate, inositol, or not at all.


Assuntos
Macrófagos/citologia , Tubo Neural/embriologia , Tubo Neural/fisiologia , Animais , Carbono/metabolismo , Linhagem da Célula , Desenvolvimento Embrionário , Ácido Fólico/metabolismo , Humanos , Modelos Teóricos , Defeitos do Tubo Neural/etiologia , Organogênese , Fenótipo , Fatores de Risco , Teratogênicos , Saco Vitelino/citologia
7.
Adv Biosyst ; 3(10): e1900064, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-32648720

RESUMO

Mechanical forces play important roles in human embryonic stem cell (hESC) differentiation. To investigate the impact of dynamic mechanical forces on neural induction of hESCs, this study employs acoustic tweezing cytometry (ATC) to apply cyclic forces/strains to hESCs by actuating integrin-bound microbubbles using ultrasound pulses. Accelerated neural induction of hESCs is demonstrated as the result of combined action of ATC and neural induction medium (NIM). Specifically, application of ATC for 30 min followed by culture in NIM upregulates neuroecdoderm markers Pax6 and Sox1 as early as 6 h after ATC, and induces neural tube-like rosette formation at 48 h after ATC. In contrast, no changes are observed in hESCs cultured in NIM without ATC treatment. In the absence of NIM, ATC application decreases Oct4, but does not increase Pax6 and Sox1 expression, nor does it induce neural rossette formation. The effects of ATC are abolished by inhibition of FAK, myosin activity, and RhoA/ROCK signaling. Taken together, the results reveal a synergistic action of ATC and NIM as an integrated mechanobiology mechanism that requires both integrin-targeted cyclic forces and chemical factors for accelerated neural induction of hESCs.


Assuntos
Fenômenos Biomecânicos/fisiologia , Células-Tronco Embrionárias Humanas , Integrinas/metabolismo , Tubo Neural , Biomarcadores/análise , Biomarcadores/metabolismo , Separação Celular , Células Cultivadas , Células-Tronco Embrionárias Humanas/citologia , Células-Tronco Embrionárias Humanas/metabolismo , Células-Tronco Embrionárias Humanas/fisiologia , Humanos , Placa Neural/citologia , Tubo Neural/citologia , Tubo Neural/metabolismo , Tubo Neural/fisiologia
8.
J Biol Chem ; 294(8): 2924-2934, 2019 02 22.
Artigo em Inglês | MEDLINE | ID: mdl-30573686

RESUMO

Neural tube closure requires apical constriction during which contraction of the apical F-actin network forces the cell into a wedged shape, facilitating the folding of the neural plate into a tube. However, how F-actin assembly at the apical surface is regulated in mammalian neurulation remains largely unknown. We report here that formin homology 2 domain-containing 3 (Fhod3), a formin protein that mediates F-actin assembly, is essential for cranial neural tube closure in mouse embryos. We found that Fhod3 is expressed in the lateral neural plate but not in the floor region of the closing neural plate at the hindbrain. Consistently, in Fhod3-null embryos, neural plate bending at the midline occurred normally, but lateral plates seemed floppy and failed to flex dorsomedially. Because the apical accumulation of F-actin and constriction were impaired specifically at the lateral plates in Fhod3-null embryos, we concluded that Fhod3-mediated actin assembly contributes to lateral plate-specific apical constriction to advance closure. Intriguingly, Fhod3 expression at the hindbrain was restricted to neuromeric segments called rhombomeres. The rhombomere-specific accumulation of apical F-actin induced by the rhombomere-restricted expression of Fhod3 was responsible for the outward bulging of rhombomeres involving apical constriction along the anteroposterior axis, as rhombomeric bulging was less prominent in Fhod3-null embryos than in the wild type. Fhod3 thus plays a crucial role in the morphological changes associated with neural tube closure at the hindbrain by mediating apical constriction not only in the mediolateral but also in the anteroposterior direction, thereby contributing to tube closure and rhombomere segmentation, respectively.


Assuntos
Proteínas dos Microfilamentos/fisiologia , Morfogênese , Placa Neural/citologia , Tubo Neural/citologia , Neurulação , Citoesqueleto de Actina/metabolismo , Animais , Células Cultivadas , Feminino , Forminas , Camundongos , Camundongos Knockout , Placa Neural/fisiologia , Tubo Neural/fisiologia
9.
Methods Mol Biol ; 1863: 47-63, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30324592

RESUMO

Developmental processes are inherently dynamic and understanding them requires quantitative measurements of gene and protein expression levels in space and time. While live imaging is a powerful approach for obtaining such data, it is still a challenge to apply it over long periods of time to large tissues, such as the embryonic spinal cord in mouse and chick. Nevertheless, dynamics of gene expression and signaling activity patterns in this organ can be studied by collecting tissue sections at different developmental stages. In combination with immunohistochemistry, this allows for measuring the levels of multiple developmental regulators in a quantitative manner with high spatiotemporal resolution. The mean protein expression levels over time, as well as embryo-to-embryo variability can be analyzed. A key aspect of the approach is the ability to compare protein levels across different samples. This requires a number of considerations in sample preparation, imaging and data analysis. Here we present a protocol for obtaining time course data of dorsoventral expression patterns from mouse and chick neural tube in the first 3 days of neural tube development. The described workflow starts from embryo dissection and ends with a processed dataset. Software scripts for data analysis are included. The protocol is adaptable and instructions that allow the user to modify different steps are provided. Thus, the procedure can be altered for analysis of time-lapse images and applied to systems other than the neural tube.


Assuntos
Embrião de Mamíferos/citologia , Regulação da Expressão Gênica no Desenvolvimento , Morfogênese , Tubo Neural/embriologia , Neurogênese , Imagem com Lapso de Tempo/métodos , Animais , Diferenciação Celular , Galinhas , Embrião de Mamíferos/fisiologia , Desenvolvimento Embrionário , Processamento de Imagem Assistida por Computador , Camundongos , Tubo Neural/crescimento & desenvolvimento , Tubo Neural/fisiologia , Transdução de Sinais
10.
Hist Philos Life Sci ; 40(3): 55, 2018 Aug 29.
Artigo em Inglês | MEDLINE | ID: mdl-30159859

RESUMO

The now classical idea that programmed cell death (apoptosis) contributes to a plethora of developmental processes still has lost nothing of its impact. It is, therefore, important to establish effective three-dimensional (3D) reconstruction as well as simulation techniques to decipher the exact patterns and functions of such apoptotic events. The present study focuses on the question whether and how apoptosis promotes neurulation-associated processes in the spinal cord of Tupaia belangeri (Tupaiidae, Scandentia, Mammalia). Our 3D reconstructions demonstrate that at least two craniocaudal waves of apoptosis consecutively pass through the dorsal spinal cord. The first wave appears to be involved in neural fold fusion and/or in selection processes among premigratory neural crest cells. The second one seems to assist in establishing the dorsal signaling center known as the roof plate. In the hindbrain, in contrast, apoptosis among premigratory neural crest cells progresses craniocaudally but discontinuously, in a segment-specific manner. Unlike apoptosis in the spinal cord, these segment-specific apoptotic events, however, precede later ones that seemingly support neural fold fusion and/or postfusion remodeling. Arguing with Whitehead that biological patterns and rhythms differ in that biological rhythms depend "upon the differences involved in each exhibition of the pattern" (Whitehead in An enquiry concerning the principles of natural knowledge. Cambridge University Press, London, 1919, p. 198) we show that 3D reconstruction and simulation techniques can contribute to distinguish between (static) patterns and (dynamic) rhythms of apoptosis. By deciphering novel patterns and rhythms of developmental apoptosis, our reconstructions help to reconcile seemingly inconsistent earlier findings in chick and mouse embryos, and to create rules for computer simulations.


Assuntos
Apoptose , Tubo Neural/fisiologia , Neurulação , Medula Espinal/fisiologia , Tupaia/fisiologia , Animais , Embrião de Mamíferos/embriologia , Imageamento Tridimensional , Tubo Neural/embriologia , Medula Espinal/embriologia , Tupaia/embriologia
11.
J Neurosci ; 38(20): 4762-4773, 2018 05 16.
Artigo em Inglês | MEDLINE | ID: mdl-29712790

RESUMO

Failure of neural tube closure leads to neural tube defects (NTDs), which can have serious neurological consequences or be lethal. Use of antiepileptic drugs (AEDs) during pregnancy increases the incidence of NTDs in offspring by unknown mechanisms. Here we show that during Xenopus laevis neural tube formation, neural plate cells exhibit spontaneous calcium dynamics that are partially mediated by glutamate signaling. We demonstrate that NMDA receptors are important for the formation of the neural tube and that the loss of their function induces an increase in neural plate cell proliferation and impairs neural cell migration, which result in NTDs. We present evidence that the AED valproic acid perturbs glutamate signaling, leading to NTDs that are rescued with varied efficacy by preventing DNA synthesis, activating NMDA receptors, or recruiting the NMDA receptor target ERK1/2. These findings may prompt mechanistic identification of AEDs that do not interfere with neural tube formation.SIGNIFICANCE STATEMENT Neural tube defects are one of the most common birth defects. Clinical investigations have determined that the use of antiepileptic drugs during pregnancy increases the incidence of these defects in the offspring by unknown mechanisms. This study discovers that glutamate signaling regulates neural plate cell proliferation and oriented migration and is necessary for neural tube formation. We demonstrate that the widely used antiepileptic drug valproic acid interferes with glutamate signaling and consequently induces neural tube defects, challenging the current hypotheses arguing that they are side effects of this antiepileptic drug that cause the increased incidence of these defects. Understanding the mechanisms of neurotransmitter signaling during neural tube formation may contribute to the identification and development of antiepileptic drugs that are safer during pregnancy.


Assuntos
Anticonvulsivantes/toxicidade , Defeitos do Tubo Neural/fisiopatologia , Tubo Neural/fisiologia , Receptores de N-Metil-D-Aspartato/fisiologia , Transdução de Sinais/fisiologia , Animais , Sinalização do Cálcio/efeitos dos fármacos , Sinalização do Cálcio/fisiologia , Movimento Celular , Proliferação de Células , Feminino , Glutamatos/fisiologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Placa Neural/citologia , Placa Neural/crescimento & desenvolvimento , Tubo Neural/crescimento & desenvolvimento , Defeitos do Tubo Neural/induzido quimicamente , Transdução de Sinais/efeitos dos fármacos , Ácido Valproico/toxicidade , Xenopus laevis
12.
PLoS Comput Biol ; 14(2): e1006003, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29470492

RESUMO

Gene regulatory networks (GRNs) control cellular function and decision making during tissue development and homeostasis. Mathematical tools based on dynamical systems theory are often used to model these networks, but the size and complexity of these models mean that their behaviour is not always intuitive and the underlying mechanisms can be difficult to decipher. For this reason, methods that simplify and aid exploration of complex networks are necessary. To this end we develop a broadly applicable form of the Zwanzig-Mori projection. By first converting a thermodynamic state ensemble model of gene regulation into mass action reactions we derive a general method that produces a set of time evolution equations for a subset of components of a network. The influence of the rest of the network, the bulk, is captured by memory functions that describe how the subnetwork reacts to its own past state via components in the bulk. These memory functions provide probes of near-steady state dynamics, revealing information not easily accessible otherwise. We illustrate the method on a simple cross-repressive transcriptional motif to show that memory functions not only simplify the analysis of the subnetwork but also have a natural interpretation. We then apply the approach to a GRN from the vertebrate neural tube, a well characterised developmental transcriptional network composed of four interacting transcription factors. The memory functions reveal the function of specific links within the neural tube network and identify features of the regulatory structure that specifically increase the robustness of the network to initial conditions. Taken together, the study provides evidence that Zwanzig-Mori projections offer powerful and effective tools for simplifying and exploring the behaviour of GRNs.


Assuntos
Regulação da Expressão Gênica , Redes Reguladoras de Genes , Memória , Tubo Neural/fisiologia , Algoritmos , Motivos de Aminoácidos , Animais , DNA/química , Camundongos , Modelos Genéticos , Modelos Estatísticos , Redes Neurais de Computação , Neurônios/fisiologia , Dinâmica não Linear , Fator de Transcrição 2 de Oligodendrócitos/metabolismo , Fator de Transcrição PAX6/metabolismo , Processos Estocásticos , Biologia de Sistemas , Termodinâmica
13.
J Neurosci Methods ; 298: 16-23, 2018 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-29408391

RESUMO

BACKGROUND: Transplantation of human pluripotent stem cell (hPSC)-derived neurons into chick embryos is an established preliminary assay to evaluate engraftment potential. Yet, with recent advances in deriving diverse human neuronal subtypes, optimizing and standardizing such transplantation methodology for specific subtypes at their correlated anatomical sites is still required. NEW METHOD: We determined the optimal stage of hPSC-derived motor neuron (hMN) differentiation for ex ovo transplantation, and developed a single injection protocol that implants hMNs throughout the spinal cord enabling broad regional engraftment possibilities. RESULTS: A single injection into the neural tube lumen yielded a 100% chick embryo survival and successful transplantation rate with MN engraftment observed from the rostral cervical through caudal lumbar spinal cord. Transplantation of HB9+/ChAT- hMN precursors yielded the greatest amount of engraftment compared to Pax6+/Nkx6.1+/Olig2+ progenitors or mature HB9+/ChAT+ hMNs. COMPARISON WITH EXISTING METHOD(S): Our single injection hMN transplant method is the first to standardize the optimal hMN phenotype for chick embryo transplantation, provide a rubric for engraftment quantification, and enable broad engraftment throughout the spinal cord with a single surgical intervention. CONCLUSION: Transplantation of HB9+/ChAT- hMN precursors into chick embryos of Hamburger Hamilton (HH) stages 15-18 using a single luminal injection confers a high probability of embryo survival and cell engraftment in diverse regions throughout the spinal cord.


Assuntos
Neurônios Motores/fisiologia , Neurônios Motores/transplante , Tubo Neural/fisiologia , Tubo Neural/cirurgia , Células-Tronco Pluripotentes/fisiologia , Células-Tronco Pluripotentes/transplante , Animais , Linhagem Celular , Embrião de Galinha , Humanos , Modelos Animais , Neurônios Motores/citologia , Tubo Neural/citologia , Neurogênese , Células-Tronco Pluripotentes/citologia , Medula Espinal/citologia , Medula Espinal/embriologia , Medula Espinal/fisiologia , Transplante Heterólogo/métodos
14.
Dev Biol ; 435(2): 130-137, 2018 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-29397878

RESUMO

Failure of neural tube closure leads to neural tube defects (NTDs), common congenital abnormalities in humans. Among the genes whose loss of function causes NTDs in mice, Grainyhead-like3 (Grhl3) is essential for spinal neural tube closure, with null mutants exhibiting fully penetrant spina bifida. During spinal neurulation Grhl3 is initially expressed in the surface (non-neural) ectoderm, subsequently in the neuroepithelial component of the neural folds and at the node-streak border, and finally in the hindgut endoderm. Here, we show that endoderm-specific knockout of Grhl3 causes late-arising spinal NTDs, preceded by increased ventral curvature of the caudal region which was shown previously to suppress closure of the spinal neural folds. This finding supports the hypothesis that diminished Grhl3 expression in the hindgut is the cause of spinal NTDs in the curly tail, carrying a hypomorphic Grhl3 allele. Complete loss of Grhl3 function produces a more severe phenotype in which closure fails earlier in neurulation, before the stage of onset of expression in the hindgut of wild-type embryos. This implicates additional tissues and NTD mechanisms in Grhl3 null embryos. Conditional knockout of Grhl3 in the neural plate and node-streak border has minimal effect on closure, suggesting that abnormal function of surface ectoderm, where Grhl3 transcripts are first detected, is primarily responsible for early failure of spinal neurulation in Grhl3 null embryos.


Assuntos
Proteínas de Ligação a DNA/fisiologia , Defeitos do Tubo Neural/genética , Tubo Neural/fisiologia , Neurulação/genética , Fatores de Transcrição/fisiologia , Animais , Proteínas de Ligação a DNA/deficiência , Proteínas de Ligação a DNA/genética , Desenvolvimento Embrionário , Regulação da Expressão Gênica no Desenvolvimento , Genes Reporter , Camadas Germinativas/metabolismo , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Placa Neural/metabolismo , Defeitos do Tubo Neural/embriologia , Defeitos do Tubo Neural/patologia , Especificidade de Órgãos , RNA Mensageiro/biossíntese , Disrafismo Espinal/embriologia , Disrafismo Espinal/genética , Fatores de Transcrição/deficiência , Fatores de Transcrição/genética
15.
Dev Biol ; 432(1): 178-191, 2017 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-28986144

RESUMO

In the spinal cord, motor axons project out the neural tube at specific exit points, then bundle together to project toward target muscles. The molecular signals that guide motor axons to and out of their exit points remain undefined. Since motor axons and their exit points are located near the floor plate, guidance signals produced by the floor plate and adjacent ventral tissues could influence motor axons as they project toward and out of exit points. The secreted Slit proteins are major floor plate repellents, and motor neurons express two Slit receptors, Robo1 and Robo2. Using mutant mouse embryos at early stages of motor axon exit, we found that motor exit points shifted ventrally in Robo1/2 or Slit1/2 double mutants. Along with the ventral shift, mutant axons had abnormal trajectories both within the neural tube toward the exit point, and after exit into the periphery. In contrast, the absence of the major ventral attractant, Netrin-1, or its receptor, DCC caused motor exit points to shift dorsally. Netrin-1 attraction on spinal motor axons was demonstrated by in vitro explant assays, showing that Netrin-1 increased outgrowth and attracted cultured spinal motor axons. The opposing effects of Slit/Robo and Netrin-1/DCC signals were tested genetically by combining Netrin-1 and Robo1/2 mutations. The location of exit points in the combined mutants was significantly recovered to their normal position compared to Netrin-1 or Robo1/2 mutants. Together, these results suggest that the proper position of motor exit points is determined by a "push-pull" mechanism, pulled ventrally by Netrin-1/DCC attraction and pushed dorsally by Slit/Robo repulsion.


Assuntos
Axônios/fisiologia , Glicoproteínas/fisiologia , Neurônios Motores/fisiologia , Proteínas do Tecido Nervoso/fisiologia , Netrinas/fisiologia , Medula Espinal/fisiologia , Animais , Axônios/metabolismo , Movimento Celular/fisiologia , Receptor DCC/metabolismo , Camundongos , Neurônios Motores/citologia , Neurônios Motores/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Netrinas/metabolismo , Tubo Neural/citologia , Tubo Neural/metabolismo , Tubo Neural/fisiologia , Receptores Imunológicos/genética , Receptores Imunológicos/metabolismo , Transdução de Sinais/genética , Medula Espinal/citologia , Medula Espinal/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Proteínas Roundabout
16.
Birth Defects Res ; 109(19): 1596-1604, 2017 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-28786179

RESUMO

BACKGROUND: Neural tube defects (NTDs) are among the most common and severe congenital malformations of the central nervous system. Animal studies have shown that apoptosis is involved in the development of NTDs. However, little evidence is available from human studies. We aim to examine the level of apoptosis and expression of apoptosis-regulating proteins of human terminated fetuses. METHODS: A total of 37 NTD cases and 21 controls from pregnancy terminations were recruited. Tissues of the central nervous system were obtained through autopsy. Apoptosis of neuroepithelial cells was examined by terminal deoxynucleotidyl transferase-mediated deoxyuridinetriphosphate nick end-labeling (TUNEL) assay. Expression of PAX3, p53, and caspase 3/8/9 in central nervous tissue was measured using Western blotting. RESULTS: More TUNEL-positive apoptosis cells were observed in the central nervous tissue of NTD cases than those of controls (p < 0.05). In spinal cord tissue, lower PAX3 expression, higher p53 expression, and increased levels of cleaved caspase 3(17kD) and cleaved caspase 8 (18kD) were found in anencephaly cases but not in spina bifida cases when compared with controls. In brain tissue, levels of PAX3 were significantly reduced in both encephalocele and spina bifida subtypes; the expression levels of cleaved caspase 3(17 kD) of encephalocele cases and cleaved caspase 8(47/45 kD) in spina bifida cases were higher than in controls; no difference was found in the expression of p53 or caspase 9 between NTDs and controls. CONCLUSION: These findings provide some evidence that excessive apoptosis in fetal central nervous tissues may be associated with the development of human NTDs. Birth Defects Research 109:1596-1604, 2017. © 2017 Wiley Periodicals, Inc.


Assuntos
Apoptose/fisiologia , Caspases/metabolismo , Defeitos do Tubo Neural/fisiopatologia , Fator de Transcrição PAX3/biossíntese , Proteína Supressora de Tumor p53/biossíntese , Feto Abortado/patologia , Aborto Induzido , Anencefalia/embriologia , Animais , Estudos de Casos e Controles , Caspases/análise , Feminino , Humanos , Marcação In Situ das Extremidades Cortadas , Lactente , Recém-Nascido , Masculino , Camundongos , Tubo Neural/metabolismo , Tubo Neural/fisiologia , Defeitos do Tubo Neural/genética , Defeitos do Tubo Neural/metabolismo , Fator de Transcrição PAX3/genética , Fator de Transcrição PAX3/metabolismo , Gravidez , Cuidado Pré-Natal , Diagnóstico Pré-Natal , Medula Espinal/patologia , Disrafismo Espinal/embriologia , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo
17.
Dev Biol ; 432(1): 24-33, 2017 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-28625870

RESUMO

Transcription factors are key orchestrators of the emergence of neuronal diversity within the developing spinal cord. As such, the two paralogous proteins Pax3 and Pax7 regulate the specification of progenitor cells within the intermediate neural tube, by defining a neat segregation between those fated to form motor circuits and those involved in the integration of sensory inputs. To attain insights into the molecular means by which they control this process, we have performed detailed phenotypic analyses of the intermediate spinal interneurons (IN), namely the dI6, V0D, V0VCG and V1 populations in compound null mutants for Pax3 and Pax7. This has revealed that the levels of Pax3/7 proteins determine both the dorso-ventral extent and the number of cells produced in each subpopulation; with increasing levels leading to the dorsalisation of their fate. Furthermore, thanks to the examination of mutants in which Pax3 transcriptional activity is skewed either towards repression or activation, we demonstrate that this cell diversification process is mainly dictated by Pax3/7 ability to repress gene expression. Consistently, we show that Pax3 and Pax7 inhibit the expression of Dbx1 and of its repressor Prdm12, fate determinants of the V0 and V1 interneurons, respectively. Notably, we provide evidence for the activity of several cis-regulatory modules of Dbx1 to be sensitive to Pax3 and Pax7 transcriptional activity levels. Altogether, our study provides insights into how the redundancy within a TF family, together with discrete dynamics of expression profiles of each member, are exploited to generate cellular diversity. Furthermore, our data supports the model whereby cell fate choices in the neural tube do not rely on binary decisions but rather on inhibition of multiple alternative fates.


Assuntos
Proteínas de Homeodomínio/fisiologia , Interneurônios/fisiologia , Proteínas do Tecido Nervoso/fisiologia , Fator de Transcrição PAX3/fisiologia , Fator de Transcrição PAX7/fisiologia , Medula Espinal/citologia , Animais , Diferenciação Celular/fisiologia , Embrião de Galinha , Regulação da Expressão Gênica no Desenvolvimento , Interneurônios/citologia , Camundongos , Tubo Neural/fisiologia , Medula Espinal/embriologia , Células-Tronco/citologia , Células-Tronco/fisiologia
18.
FASEB J ; 31(8): 3622-3635, 2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-28432198

RESUMO

Periconception maternal folic acid (vitamin B9) supplementation can reduce the prevalence of neural tube defects (NTDs), although just how folates benefit the developing embryo and promote closing of the neural tube and other morphologic processes during development remains unknown. Folate contributes to a 1-carbon metabolism, which is essential for purine biosynthesis and methionine recycling and affects methylation of DNA, histones, and nonhistone proteins. Herein, we used animal models and cultured mammalian cells to demonstrate that disruption of the methylation pathway mediated by folate compromises normal neural tube closure (NTC) and ciliogenesis. We demonstrate that the embryos with NTD failed to adequately methylate septin2, a key regulator of cilium structure and function. We report that methylation of septin2 affected its GTP binding activity and formation of the septin2-6-7 complex. We propose that folic acid promotes normal NTC in some embryos by regulating the methylation of septin2, which is critical for normal cilium formation during early embryonic development.-Toriyama, M., Toriyama, M., Wallingford, J. B., Finnell, R. H. Folate-dependent methylation of septins governs ciliogenesis during neural tube closure.


Assuntos
Cílios/fisiologia , Embrião de Mamíferos/metabolismo , Embrião não Mamífero/metabolismo , Ácido Fólico/metabolismo , Tubo Neural/fisiologia , Septinas/metabolismo , Animais , Dactinomicina/análogos & derivados , Desenvolvimento Embrionário/fisiologia , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Células HEK293 , Proteínas Hedgehog/genética , Proteínas Hedgehog/metabolismo , Humanos , Metilação , Camundongos , Defeitos do Tubo Neural/etiologia , Plasmídeos , Transdução de Sinais , Xenopus/embriologia
19.
PLoS Comput Biol ; 13(1): e1005307, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-28135279

RESUMO

A fundamental question in biology is how sharp boundaries of gene expression form precisely in spite of biological variation/noise. Numerous mechanisms position gene expression domains across fields of cells (e.g. morphogens), but how these domains are refined remains unclear. In some cases, domain boundaries sharpen through differential adhesion-mediated cell sorting. However, boundaries can also sharpen through cellular plasticity, with cell fate changes driven by up- or down-regulation of gene expression. In this context, we have argued that noise in gene expression can help cells transition to the correct fate. Here we investigate the efficacy of cell sorting, gene expression plasticity, and their combination in boundary sharpening using multi-scale, stochastic models. We focus on the formation of hindbrain segments (rhombomeres) in the developing zebrafish as an example, but the mechanisms investigated apply broadly to many tissues. Our results indicate that neither sorting nor plasticity is sufficient on its own to sharpen transition regions between different rhombomeres. Rather the two have complementary strengths and weaknesses, which synergize when combined to sharpen gene expression boundaries.


Assuntos
Regulação da Expressão Gênica/fisiologia , Modelos Biológicos , Modelos Estatísticos , Tubo Neural/citologia , Tubo Neural/fisiologia , Plasticidade Neuronal/fisiologia , Animais , Adesão Celular/fisiologia , Movimento Celular , Expressão Gênica/fisiologia , Morfogênese , Rombencéfalo/citologia , Rombencéfalo/fisiologia , Razão Sinal-Ruído , Processos Estocásticos , Peixe-Zebra
20.
Int J Dev Biol ; 61(3-4-5): 257-265, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-27528042

RESUMO

During embryonic development of the Central Nervous System (CNS), the expression of the bHLH transcription factor Nato3 (Ferd3l) is unique and restricted to the floor plate of the neural tube. In mice lacking Nato3 the floor plate cells of the spinal cord do not fully mature, whereas in the midbrain floor plate, progenitors lose some neurogenic activity, giving rise to a reduced population of dopaminergic neurons. Since the floor plate is considered to be disintegrated at the time of birth, Nato3 expression was never tested postnatally and in adult mice. Here, we utilized a Nato3 knockout mouse model in which a LacZ reporter precisely replaced the coding region under the endogenous regulatory elements, so that its expression recapitulates the spatiotemporal pattern of Nato3 expression. Nato3 was found to be expressed in the CNS throughout life in a highly restricted manner along the medial cavities: in subpopulations of cells in the IIIrd ventricle, the cerebral aqueduct, the IVth ventricle, the central canal of the spinal cord, and the subcommissural organ, a gland located in the midbrain. A few unifying themes are shared among all Nato3-positive cells: all are positioned in the midline, are of an ependymal type, and contact the cerebrospinal fluid (CSF) similarly to the embryonic position of the floor plate bordering the lumen of the neural tube. Taken together, Nato3 defines an unrecognized subpopulation of medial cells positioned at only one side of circular ependymal structures, and it may affect their regulatory activities and neuronal stem cell function.


Assuntos
Sistema Nervoso Central/fisiologia , Epêndima/fisiologia , Regulação da Expressão Gênica no Desenvolvimento , Animais , Diferenciação Celular , Líquido Cefalorraquidiano/metabolismo , Óperon Lac , Camundongos , Camundongos Knockout , Microscopia de Fluorescência , Proteínas do Tecido Nervoso/metabolismo , Tubo Neural/fisiologia , Neurogênese , Neurônios/metabolismo , Proteínas Repressoras , Medula Espinal/embriologia , Células-Tronco/metabolismo , Fatores de Tempo , Fatores de Transcrição/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA