Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 266
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Genes Dev ; 32(2): 165-180, 2018 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-29440260

RESUMO

Multiple congenital disorders often present complex phenotypes, but how the mutation of individual genetic factors can lead to multiple defects remains poorly understood. In the present study, we used human neuroepithelial (NE) cells and CHARGE patient-derived cells as an in vitro model system to identify the function of chromodomain helicase DNA-binding 7 (CHD7) in NE-neural crest bifurcation, thus revealing an etiological link between the central nervous system (CNS) and craniofacial anomalies observed in CHARGE syndrome. We found that CHD7 is required for epigenetic activation of superenhancers and CNS-specific enhancers, which support the maintenance of the NE and CNS lineage identities. Furthermore, we found that BRN2 and SOX21 are downstream effectors of CHD7, which shapes cellular identities by enhancing a CNS-specific cellular program and indirectly repressing non-CNS-specific cellular programs. Based on our results, CHD7, through its interactions with superenhancer elements, acts as a regulatory hub in the orchestration of the spatiotemporal dynamics of transcription factors to regulate NE and CNS lineage identities.


Assuntos
DNA Helicases/fisiologia , Proteínas de Ligação a DNA/fisiologia , Epigênese Genética , Células-Tronco Neurais/metabolismo , Células Neuroepiteliais/metabolismo , Síndrome CHARGE/genética , Linhagem Celular , Linhagem da Célula/genética , DNA Helicases/genética , DNA Helicases/metabolismo , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Elementos Facilitadores Genéticos , Haploinsuficiência , Humanos , Crista Neural/metabolismo , Transcrição Gênica
2.
J Cell Sci ; 135(6)2022 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-35217862

RESUMO

Dynamic contacts between cells within the developing neuroepithelium are poorly understood but play important roles in cell and tissue morphology and cell signalling. Here, using live-cell imaging and electron microscopy we reveal multiple protrusive structures in neuroepithelial apical endfeet of the chick embryonic spinal cord, including sub-apical protrusions that extend laterally within the tissue, and observe similar structures in human neuroepithelium. We characterise the dynamics, shape and cytoskeleton of these lateral protrusions and distinguish them from cytonemes, filopodia and tunnelling nanotubes. We demonstrate that lateral protrusions form a latticework of membrane contacts between non-adjacent cells, depend on actin but not microtubule dynamics, and provide a lamellipodial-like platform for further extending fine actin-dependent filipodia. We find that lateral protrusions depend on the actin-binding protein WAVE1 (also known as WASF1): misexpression of mutant WAVE1 attenuated protrusion and generated a round-ended apical endfoot morphology. However, this did not alter apico-basal cell polarity or tissue integrity. During normal neuronal delamination, lateral protrusions were withdrawn, but precocious protrusion loss induced by mutant WAVE1 was insufficient to trigger neurogenesis. This study uncovers a new form of cell-cell contact within the developing neuroepithelium, regulation of which prefigures neuronal delamination. This article has an associated First Person interview with the first author of the paper.


Assuntos
Actinas , Células Neuroepiteliais , Actinas/metabolismo , Citoesqueleto/metabolismo , Humanos , Células Neuroepiteliais/metabolismo , Neurogênese , Pseudópodes/metabolismo , Família de Proteínas da Síndrome de Wiskott-Aldrich/metabolismo
3.
Development ; 148(2)2021 01 26.
Artigo em Inglês | MEDLINE | ID: mdl-33500317

RESUMO

Pathogenic mutations in the endocytic receptor LRP2 in humans are associated with severe neural tube closure defects (NTDs) such as anencephaly and spina bifida. Here, we have combined analysis of neural tube closure in mouse and in the African Clawed Frog Xenopus laevis to elucidate the etiology of Lrp2-related NTDs. Lrp2 loss of function impaired neuroepithelial morphogenesis, culminating in NTDs that impeded anterior neural plate folding and neural tube closure in both model organisms. Loss of Lrp2 severely affected apical constriction as well as proper localization of the core planar cell polarity (PCP) protein Vangl2, demonstrating a highly conserved role of the receptor in these processes, which are essential for neural tube formation. In addition, we identified a novel functional interaction of Lrp2 with the intracellular adaptor proteins Shroom3 and Gipc1 in the developing forebrain. Our data suggest that, during neurulation, motifs within the intracellular domain of Lrp2 function as a hub that orchestrates endocytic membrane removal for efficient apical constriction, as well as PCP component trafficking in a temporospatial manner.


Assuntos
Endocitose , Espaço Intracelular/metabolismo , Proteína-2 Relacionada a Receptor de Lipoproteína de Baixa Densidade/metabolismo , Tubo Neural/embriologia , Animais , Membrana Celular/metabolismo , Polaridade Celular , Proteína-2 Relacionada a Receptor de Lipoproteína de Baixa Densidade/deficiência , Camundongos Endogâmicos C57BL , Modelos Biológicos , Morfogênese , Tubo Neural/metabolismo , Tubo Neural/ultraestrutura , Células Neuroepiteliais/metabolismo , Prosencéfalo/metabolismo , Ligação Proteica , Xenopus , Proteínas de Xenopus/metabolismo
4.
Development ; 148(17)2021 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-34463328

RESUMO

Pathogenic gene variants in humans that affect the sonic hedgehog (SHH) pathway lead to severe brain malformations with variable penetrance due to unknown modifier genes. To identify such modifiers, we established novel congenic mouse models. LRP2-deficient C57BL/6N mice suffer from heart outflow tract defects and holoprosencephaly caused by impaired SHH activity. These defects are fully rescued on a FVB/N background, indicating a strong influence of modifier genes. Applying comparative transcriptomics, we identified Pttg1 and Ulk4 as candidate modifiers upregulated in the rescue strain. Functional analyses showed that ULK4 and PTTG1, both microtubule-associated proteins, are positive regulators of SHH signaling, rendering the pathway more resilient to disturbances. In addition, we characterized ULK4 and PTTG1 as previously unidentified components of primary cilia in the neuroepithelium. The identification of genes that powerfully modulate the penetrance of genetic disturbances affecting the brain and heart is likely relevant to understanding the variability in human congenital disorders.


Assuntos
Encéfalo/embriologia , Genes Modificadores/fisiologia , Proteínas Hedgehog/metabolismo , Transdução de Sinais , Animais , Encéfalo/metabolismo , Cílios/metabolismo , Modelos Animais de Doenças , Cardiopatias Congênitas/genética , Proteínas Hedgehog/genética , Holoprosencefalia/genética , Proteína-2 Relacionada a Receptor de Lipoproteína de Baixa Densidade/genética , Proteína-2 Relacionada a Receptor de Lipoproteína de Baixa Densidade/metabolismo , Camundongos , Mutação , Células Neuroepiteliais/metabolismo , Penetrância , Fenótipo , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Securina/genética , Securina/metabolismo
5.
Cell ; 138(6): 1195-208, 2009 Sep 18.
Artigo em Inglês | MEDLINE | ID: mdl-19766571

RESUMO

Progenitor cell nuclei in the rapidly expanding epithelium of the embryonic vertebrate central nervous system undergo a process called interkinetic nuclear migration (IKNM). Movements of IKNM are generally believed to involve smooth migration of nuclei from apical to basal and back during the G1 and G2 phases of the cell cycle, respectively. Yet, this has not been formally demonstrated, nor have the molecular mechanisms that drive IKNM been identified. Using time-lapse confocal microscopy to observe nuclear movements in zebrafish retinal neuroepithelial cells, we show that, except for brief apical nuclear translocations preceding mitosis, IKNM is stochastic rather than smooth and directed. We also show that IKNM is driven largely by actomyosin-dependent forces as it still occurs when the microtubule cytoskeleton is compromised but is blocked when MyosinII activity is inhibited.


Assuntos
Actomiosina/metabolismo , Núcleo Celular/metabolismo , Retina/citologia , Peixe-Zebra/embriologia , Animais , Complexo Dinactina , Embrião não Mamífero/citologia , Embrião não Mamífero/metabolismo , Proteínas Associadas aos Microtúbulos/metabolismo , Células Neuroepiteliais/citologia , Células Neuroepiteliais/metabolismo , Retina/embriologia , Peixe-Zebra/metabolismo , Proteínas de Peixe-Zebra/metabolismo
6.
J Exp Biol ; 226(8)2023 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-37097020

RESUMO

The study of breathing in fishes has featured prominently in Journal of Experimental Biology (JEB), particularly during the latter half of the past century. Indeed, many of the seminal discoveries in this important sub-field of comparative respiratory physiology were reported first in JEB. The period spanning 1960-1990 (the 'golden age of comparative respiratory physiology') witnessed intense innovation in the development of methods to study the control of breathing. Many of the guiding principles of piscine ventilatory control originated during this period, including our understanding of the dominance of O2 as the driver of ventilation in fish. However, a critical issue - the identity of the peripheral O2 chemoreceptors - remained unanswered until methods for cell isolation, culture and patch-clamp recording established that gill neuroepithelial cells (NECs) respond to hypoxia in vitro. Yet, the role of the NECs and other putative peripheral or central chemoreceptors in the control of ventilation in vivo remains poorly understood. Further progress will be driven by the implementation of genetic tools, most of which can be used in zebrafish (Danio rerio). These tools include CRISPR/Cas9 for selective gene knockout, and Tol2 systems for transgenesis, the latter of which enables optogenetic stimulation of cellular pathways, cellular ablation and in vivo cell-specific biosensing. Using these methods, the next period of discovery will see the identification of the peripheral sensory pathways that initiate ventilatory responses, and will elucidate the nature of their integration within the central nervous system and their link to the efferent motor neurons that control breathing.


Assuntos
Oxigênio , Peixe-Zebra , Animais , Peixe-Zebra/fisiologia , Oxigênio/metabolismo , Peixes/fisiologia , Células Neuroepiteliais/metabolismo , Células Quimiorreceptoras/metabolismo , Respiração , Brânquias/metabolismo
7.
Cell ; 134(6): 1055-65, 2008 Sep 19.
Artigo em Inglês | MEDLINE | ID: mdl-18805097

RESUMO

The different cell types in the central nervous system develop from a common pool of progenitor cells. The nuclei of progenitors move between the apical and basal surfaces of the neuroepithelium in phase with their cell cycle, a process termed interkinetic nuclear migration (INM). In the retina of zebrafish mikre oko (mok) mutants, in which the motor protein Dynactin-1 is disrupted, interkinetic nuclei migrate more rapidly and deeply to the basal side and more slowly to the apical side. We found that Notch signaling is predominantly activated on the apical side in both mutants and wild-type. Mutant progenitors are, thus, less exposed to Notch and exit the cell cycle prematurely. This leads to an overproduction of early-born retinal ganglion cells (RGCs) at the expense of later-born interneurons and glia. Our data indicate that the function of INM is to balance the exposure of progenitor nuclei to neurogenic versus proliferative signals.


Assuntos
Núcleo Celular/metabolismo , Células Neuroepiteliais/citologia , Organogênese , Retina/embriologia , Animais , Padronização Corporal , Ciclo Celular , Diferenciação Celular , Complexo Dinactina , Embrião não Mamífero/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Proteínas Associadas aos Microtúbulos/genética , Mutação , Células Neuroepiteliais/metabolismo , Receptores Notch/metabolismo , Retina/citologia , Células Ganglionares da Retina/metabolismo , Células-Tronco/citologia , Células-Tronco/metabolismo , Peixe-Zebra , Proteínas de Peixe-Zebra/genética
8.
Dev Biol ; 478: 59-75, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34029538

RESUMO

Morphogenesis of the vertebrate neural tube occurs by elongation and bending of the neural plate, tissue shape changes that are driven at the cellular level by polarized cell intercalation and cell shape changes, notably apical constriction and cell wedging. Coordinated cell intercalation, apical constriction, and wedging undoubtedly require complex underlying cytoskeletal dynamics and remodeling of adhesions. Mutations of the gene encoding Scribble result in neural tube defects in mice, however the cellular and molecular mechanisms by which Scrib regulates neural cell behavior remain unknown. Analysis of Scribble mutants revealed defects in neural tissue shape changes, and live cell imaging of mouse embryos showed that the Scrib mutation results in defects in polarized cell intercalation, particularly in rosette resolution, and failure of both cell apical constriction and cell wedging. Scrib mutant embryos displayed aberrant expression of the junctional proteins ZO-1, Par3, Par6, E- and N-cadherins, and the cytoskeletal proteins actin and myosin. These findings show that Scribble has a central role in organizing the molecular complexes regulating the morphomechanical neural cell behaviors underlying vertebrate neurulation, and they advance our understanding of the molecular mechanisms involved in mammalian neural tube closure.


Assuntos
Peptídeos e Proteínas de Sinalização Intracelular/genética , Defeitos do Tubo Neural/embriologia , Tubo Neural/embriologia , Animais , Polaridade Celular , Forma Celular , Proteínas do Citoesqueleto , Expressão Gênica , Junções Intercelulares/metabolismo , Junções Intercelulares/ultraestrutura , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Camundongos , Morfogênese , Mutação , Proteínas do Tecido Nervoso/genética , Placa Neural/citologia , Placa Neural/embriologia , Tubo Neural/citologia , Defeitos do Tubo Neural/genética , Células Neuroepiteliais/citologia , Células Neuroepiteliais/metabolismo , Células Neuroepiteliais/ultraestrutura , Proteínas de Junções Íntimas/genética , Proteínas de Junções Íntimas/metabolismo
9.
Development ; 146(20)2019 10 24.
Artigo em Inglês | MEDLINE | ID: mdl-31575649

RESUMO

Massive, coordinated cellular changes accompany the transition of central nervous system (CNS) progenitors from forebrain neurectodermal cells to specified neuroepithelial cells. We have previously found that MYC regulates the changing ribosomal and proteostatic landscapes in mouse forebrain precursors at embryonic days E8.5 and E10.5 (before and after neural tube closure; NTC) (Chau et al., 2018). Here, we demonstrate parallel coordinated transcriptional changes in metabolic machinery during this same stage of forebrain specification. Progenitors showed striking mitochondrial structural changes transitioning from glycolytic cristae at E8.5, to more traditional mitochondria at E10.5. Accordingly, glucose use shifted in progenitors such that E8.5 progenitors relied on glycolysis, and after NTC increasingly used oxidative phosphorylation. This metabolic shift was matched by changes in surrounding amniotic and cerebrospinal fluid proteomes. Importantly, these mitochondrial morphological shifts depend on MYC downregulation. Together, our findings demonstrate that metabolic shifting accompanies dynamic organelle and proteostatic remodeling of progenitor cells during the earliest stages of forebrain development.


Assuntos
Mitocôndrias/metabolismo , Proteoma/metabolismo , Animais , Sistema Nervoso Central/metabolismo , Epitélio/metabolismo , Feminino , Glicólise , Immunoblotting , Masculino , Camundongos , Camundongos Mutantes , Microscopia Eletrônica de Transmissão , Células Neuroepiteliais/citologia , Células Neuroepiteliais/metabolismo , Prosencéfalo/citologia , Prosencéfalo/metabolismo , RNA-Seq , Reação em Cadeia da Polimerase Via Transcriptase Reversa
10.
J Cell Sci ; 132(13)2019 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-31182644

RESUMO

Cellular generation of mechanical forces required to close the presumptive spinal neural tube, the 'posterior neuropore' (PNP), involves interkinetic nuclear migration (INM) and apical constriction. Both processes change the apical surface area of neuroepithelial cells, but how they are biomechanically integrated is unknown. Rho kinase (Rock; herein referring to both ROCK1 and ROCK2) inhibition in mouse whole embryo culture progressively widens the PNP. PNP widening is not caused by increased mechanical tension opposing closure, as evidenced by diminished recoil following laser ablation. Rather, Rock inhibition diminishes neuroepithelial apical constriction, producing increased apical areas in neuroepithelial cells despite diminished tension. Neuroepithelial apices are also dynamically related to INM progression, with the smallest dimensions achieved in cells positive for the pan-M phase marker Rb phosphorylated at S780 (pRB-S780). A brief (2 h) Rock inhibition selectively increases the apical area of pRB-S780-positive cells, but not pre-anaphase cells positive for phosphorylated histone 3 (pHH3+). Longer inhibition (8 h, more than one cell cycle) increases apical areas in pHH3+ cells, suggesting cell cycle-dependent accumulation of cells with larger apical surfaces during PNP widening. Consequently, arresting cell cycle progression with hydroxyurea prevents PNP widening following Rock inhibition. Thus, Rock-dependent apical constriction compensates for the PNP-widening effects of INM to enable progression of closure.This article has an associated First Person interview with the first authors of the paper.


Assuntos
Divisão Celular , Tubo Neural/citologia , Tubo Neural/metabolismo , Quinases Associadas a rho/metabolismo , Actomiosina/metabolismo , Animais , Ciclo Celular/efeitos dos fármacos , Embrião de Mamíferos/citologia , Camundongos , Células Neuroepiteliais/citologia , Células Neuroepiteliais/efeitos dos fármacos , Células Neuroepiteliais/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Quinases Associadas a rho/antagonistas & inibidores
11.
Development ; 145(2)2018 01 25.
Artigo em Inglês | MEDLINE | ID: mdl-29361557

RESUMO

Whether common principles regulate the self-renewing potential of neural stem cells (NSCs) throughout the developing central nervous system is still unclear. In the Drosophila ventral nerve cord and central brain, asymmetrically dividing NSCs, called neuroblasts (NBs), progress through a series of sequentially expressed transcription factors that limits self-renewal by silencing a genetic module involving the transcription factor Chinmo. Here, we find that Chinmo also promotes neuroepithelium growth in the optic lobe during early larval stages by boosting symmetric self-renewing divisions while preventing differentiation. Neuroepithelium differentiation in late larvae requires the transcriptional silencing of chinmo by ecdysone, the main steroid hormone, therefore allowing coordination of neural stem cell self-renewal with organismal growth. In contrast, chinmo silencing in NBs is post-transcriptional and does not require ecdysone. Thus, during Drosophila development, humoral cues or tissue-intrinsic temporal specification programs respectively limit self-renewal in different types of neural progenitors through the transcriptional and post-transcriptional regulation of the same transcription factor.


Assuntos
Proliferação de Células/fisiologia , Proteínas de Drosophila/metabolismo , Inativação Gênica/fisiologia , Proteínas do Tecido Nervoso/metabolismo , Células-Tronco Neurais/metabolismo , Células Neuroepiteliais/metabolismo , Animais , Proteínas de Drosophila/genética , Drosophila melanogaster , Ecdisona/biossíntese , Ecdisona/genética , Proteínas do Tecido Nervoso/genética , Células-Tronco Neurais/citologia , Células Neuroepiteliais/citologia
12.
Development ; 145(22)2018 11 19.
Artigo em Inglês | MEDLINE | ID: mdl-30333212

RESUMO

The folding of epithelial tissues is crucial for development of three-dimensional structure and function. Understanding this process can assist in determining the etiology of developmental disease and engineering of tissues for the future of regenerative medicine. Folding of epithelial tissues towards the apical surface has long been studied, but the molecular mechanisms that mediate epithelial folding towards the basal surface are just emerging. Here, we utilize zebrafish neuroepithelium to identify mechanisms that mediate basal tissue folding to form the highly conserved embryonic midbrain-hindbrain boundary. Live imaging revealed Wnt5b as a mediator of anisotropic epithelial cell shape, both apically and basally. In addition, we uncovered a Wnt5b-mediated mechanism for specific regulation of basal anisotropic cell shape that is microtubule dependent and likely to involve JNK signaling. We propose a model in which a single morphogen can differentially regulate apical versus basal cell shape during tissue morphogenesis.


Assuntos
Epitélio/metabolismo , Microtúbulos/metabolismo , Morfogênese , Peixe-Zebra/embriologia , Animais , Anisotropia , Forma Celular , Embrião não Mamífero/citologia , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Mesencéfalo/citologia , Mesencéfalo/embriologia , Células Neuroepiteliais/citologia , Células Neuroepiteliais/metabolismo , Polimerização , Rombencéfalo/citologia , Rombencéfalo/embriologia , Tubulina (Proteína)/metabolismo
13.
Development ; 145(19)2018 10 05.
Artigo em Inglês | MEDLINE | ID: mdl-30228103

RESUMO

Neural tube closure relies on the apical constriction of neuroepithelial cells. Research in frog and fly embryos has found links between the levels of intracellular calcium, actomyosin dynamics and apical constriction. However, genetic evidence for a role of calcium in apical constriction during mammalian neurulation is still lacking. Secretory pathway calcium ATPase (SPCA1) regulates calcium homeostasis by pumping cytosolic calcium into the Golgi apparatus. Loss of function in Spca1 causes cranial exencephaly and spinal cord defects in mice, phenotypes previously ascribed to apoptosis. However, our characterization of a novel allele of Spca1 revealed that neurulation defects in Spca1 mutants are not due to cell death, but rather to a failure of neuroepithelial cells to apically constrict. We show that SPCA1 influences cell contractility by regulating myosin II localization. Furthermore, we found that loss of Spca1 disrupts actin dynamics and the localization of the actin remodeling protein cofilin 1. Taken together, our results provide evidence that SPCA1 promotes neurulation by regulating the cytoskeletal dynamics that promote apical constriction and identify cofilin 1 as a downstream effector of SPCA1 function.


Assuntos
ATPases Transportadoras de Cálcio/metabolismo , Citoesqueleto/metabolismo , Tubo Neural/embriologia , Tubo Neural/enzimologia , Via Secretória , Citoesqueleto de Actina/metabolismo , Alelos , Sequência de Aminoácidos , Animais , Apoptose , Sequência de Bases , Cálcio/metabolismo , ATPases Transportadoras de Cálcio/genética , Cofilina 1/metabolismo , Feminino , Testes Genéticos , Homeostase , Masculino , Camundongos Endogâmicos C57BL , Mutação/genética , Miosina Tipo II/metabolismo , Células Neuroepiteliais/metabolismo , Fosforilação , Medula Espinal/embriologia , Medula Espinal/patologia
14.
Development ; 145(24)2018 12 13.
Artigo em Inglês | MEDLINE | ID: mdl-30545932

RESUMO

Micronutrition is essential for neural tube closure, and zinc deficiency is associated with human neural tube defects. Here, we modeled zinc deficiency in mouse embryos, and used live imaging and molecular studies to determine how zinc deficiency affects neural tube closure. Embryos cultured with the zinc chelator TPEN failed to close the neural tube and showed excess apoptosis. TPEN-induced p53 protein stabilization in vivo and in neuroepithelial cell cultures and apoptosis was dependent on p53. Mechanistically, zinc deficiency resulted in disrupted interaction between p53 and the zinc-dependent E3 ubiquitin ligase Mdm2, and greatly reduced p53 ubiquitylation. Overexpression of human CHIP, a zinc-independent E3 ubiquitin ligase that targets p53, relieved TPEN-induced p53 stabilization and reduced apoptosis. Expression of p53 pro-apoptotic target genes was upregulated by zinc deficiency. Correspondingly, embryos cultured with p53 transcriptional activity inhibitor pifithrin-α could overcome TPEN-induced apoptosis and failure of neural tube closure. Our studies indicate that zinc deficiency disrupts neural tube closure through decreased p53 ubiquitylation, increased p53 stabilization and excess apoptosis.


Assuntos
Defeitos do Tubo Neural/metabolismo , Defeitos do Tubo Neural/patologia , Proteína Supressora de Tumor p53/metabolismo , Ubiquitinação , Zinco/deficiência , Animais , Apoptose , Linhagem Celular , Camundongos , Mitocôndrias/metabolismo , Modelos Biológicos , Tubo Neural/anormalidades , Tubo Neural/patologia , Células Neuroepiteliais/metabolismo , Transdução de Sinais , Transcrição Gênica , Ativação Transcricional/genética
15.
Cell Tissue Res ; 384(1): 35-47, 2021 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-33404838

RESUMO

In teleost fish, specialized oxygen (O2) chemoreceptors, called neuroepithelial cells (NECs), are found in the gill epithelium in adults. During development, NECs are present in the skin before the formation of functional gills. NECs are known for retaining the monoamine neurotransmitter, serotonin (5-HT) and are conventionally identified through immunoreactivity with antibodies against 5-HT or synaptic vesicle protein (SV2). However, identification of NECs in live tissue and isolated cell preparations has been challenging due to the lack of a specific marker. The present study explored the use of the transgenic zebrafish, ETvmat2:GFP, which expresses green fluorescent protein (GFP) under the control of the vesicular monoamine transporter 2 (vmat2) regulatory element, to identify NECs. Using immunohistochemistry and confocal microscopy, we confirmed that the endogenous GFP in ETvmat2:GFP labelled serotonergic NECs in the skin of larvae and in the gills of adults. NECs of the gill filaments expressed a higher level of endogenous GFP compared with other cells. The endogenous GFP also labelled intrabranchial neurons of the gill filaments. Flow cytometric analysis demonstrated that filamental NECs could be distinguished from other dissociated gill cells based on high GFP expression alone. Acclimation to 2 weeks of severe hypoxia (PO2 = 35 mmHg) induced an increase in filamental NEC frequency, size and GFP gene expression. Here we present for the first time a transgenic tool that labels O2 chemoreceptors in an aquatic vertebrate and its use in high-throughput experimentation.


Assuntos
Genes Reporter/genética , Células Neuroepiteliais/metabolismo , Animais , Animais Geneticamente Modificados , Imuno-Histoquímica , Peixe-Zebra
16.
Neurochem Res ; 46(10): 2512-2524, 2021 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-33725233

RESUMO

Radial glial cells (RGC) are at the center of brain development in vertebrates, acting as progenitors for neurons and macroglia (oligodendrocytes and astrocytes) and as guides for migration of neurons from the ventricular surface to their final positions in the brain. These cells originate from neuroepithelial cells (NEC) from which they inherit their epithelial features and polarized morphology, with processes extending from the ventricular to the pial surface of the embryonic cerebrum. We have learnt a great deal since the first descriptions of these cells at the end of the nineteenth century. However, there are still questions regarding how and when NEC transform into RGC or about the function of intermediate filaments such as glial fibrillary acidic protein (GFAP) in RGCs and their dynamics during neurogenesis. For example, it is not clear why RGCs in primates, including humans, express GFAP at the onset of cortical neurogenesis while in rodents it is expressed when it is essentially complete. Based on an ultrastructural analysis of GFAP expression and cell morphology of dividing progenitors in the developing neocortex of the macaque monkey, we show that RGCs become the main progenitor in the developing cerebrum by the start of neurogenesis, as all dividing cells show glial features such as GFAP expression and lack of tight junctions. Also, our data suggest that RGCs retract their apical process during mitosis. We discuss our findings in the context of the role and molecular characteristics of RGCs in the vertebrate brain, their differences with NECs and their dynamic behavior during the process of neurogenesis.


Assuntos
Células Ependimogliais/metabolismo , Neurogênese/fisiologia , Animais , Ciclo Celular/fisiologia , Diferenciação Celular/fisiologia , Extensões da Superfície Celular/metabolismo , Proteína Glial Fibrilar Ácida/metabolismo , Macaca , Proteínas do Tecido Nervoso/metabolismo , Células Neuroepiteliais/metabolismo
17.
PLoS Biol ; 16(4): e2004162, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29708962

RESUMO

The vertebrate neuroepithelium is composed of elongated progenitors whose reciprocal attachments ensure the continuity of the ventricular wall. As progenitors commit to differentiation, they translocate their nucleus basally and eventually withdraw their apical endfoot from the ventricular surface. However, the mechanisms allowing this delamination process to take place while preserving the integrity of the neuroepithelial tissue are still unclear. Here, we show that Notch signaling, which is classically associated with an undifferentiated state, remains active in prospective neurons until they delaminate. During this transition period, prospective neurons rapidly reduce their apical surface and only later down-regulate N-Cadherin levels. Upon Notch blockade, nascent neurons disassemble their junctions but fail to reduce their apical surface. This disrupted sequence weakens the junctional network and eventually leads to breaches in the ventricular wall. We also provide evidence that the Notch ligand Delta-like 1 (Dll1) promotes differentiation by reducing Notch signaling through a Cis-inhibition mechanism. However, during the delamination process, the ubiquitin ligase Mindbomb1 (Mib1) transiently blocks this Cis-inhibition and sustains Notch activity to defer differentiation. We propose that the fine-tuned balance between Notch Trans-activation and Cis-inhibition allows neuroepithelial cells to seamlessly delaminate from the ventricular wall as they commit to differentiation.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Peptídeos e Proteínas de Sinalização Intracelular/genética , Proteínas de Membrana/genética , Células Neuroepiteliais/metabolismo , Neurogênese/genética , Receptores Notch/genética , Ubiquitina-Proteína Ligases/genética , Animais , Animais Geneticamente Modificados , Caderinas/genética , Caderinas/metabolismo , Diferenciação Celular , Embrião de Galinha , Galinhas , Feminino , Vetores Genéticos/química , Vetores Genéticos/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Lentivirus/genética , Lentivirus/metabolismo , Masculino , Proteínas de Membrana/metabolismo , Células Neuroepiteliais/citologia , Neurônios/citologia , Neurônios/metabolismo , Plasmídeos/química , Plasmídeos/metabolismo , Receptores Notch/metabolismo , Transdução de Sinais , Transfecção , Ubiquitina-Proteína Ligases/metabolismo
18.
PLoS Genet ; 14(4): e1007353, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29677185

RESUMO

The central nervous system develops from monolayered neuroepithelial sheets. In a first step patterning mechanisms subdivide the seemingly uniform epithelia into domains allowing an increase of neuronal diversity in a tightly controlled spatial and temporal manner. In Drosophila, neuroepithelial patterning of the embryonic optic placode gives rise to the larval eye primordium, consisting of two photoreceptor (PR) precursor types (primary and secondary), as well as the optic lobe primordium, which during larval and pupal stages develops into the prominent optic ganglia. Here, we characterize a genetic network that regulates the balance between larval eye and optic lobe precursors, as well as between primary and secondary PR precursors. In a first step the proneural factor Atonal (Ato) specifies larval eye precursors, while the orphan nuclear receptor Tailless (Tll) is crucial for the specification of optic lobe precursors. The Hedgehog and Notch signaling pathways act upstream of Ato and Tll to coordinate neural precursor specification in a timely manner. The correct spatial placement of the boundary between Ato and Tll in turn is required to control the precise number of primary and secondary PR precursors. In a second step, Notch signaling also controls a binary cell fate decision, thus, acts at the top of a cascade of transcription factor interactions to define PR subtype identity. Our model serves as an example of how combinatorial action of cell extrinsic and cell intrinsic factors control neural tissue patterning.


Assuntos
Drosophila melanogaster/crescimento & desenvolvimento , Drosophila melanogaster/genética , Olho/crescimento & desenvolvimento , Olho/metabolismo , Animais , Animais Geneticamente Modificados , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Padronização Corporal/genética , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Redes Reguladoras de Genes , Genes de Insetos , Proteínas Hedgehog/genética , Proteínas Hedgehog/metabolismo , Larva/genética , Larva/crescimento & desenvolvimento , Larva/metabolismo , Mutação , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Células Neuroepiteliais/metabolismo , Lobo Óptico de Animais não Mamíferos/crescimento & desenvolvimento , Lobo Óptico de Animais não Mamíferos/metabolismo , Células Fotorreceptoras de Invertebrados/citologia , Células Fotorreceptoras de Invertebrados/metabolismo , Receptores Notch/genética , Receptores Notch/metabolismo , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Transdução de Sinais
19.
Artigo em Inglês | MEDLINE | ID: mdl-33444774

RESUMO

Ammonia is both a respiratory gas and a toxicant in teleost fish. Hyperventilation is a well-known response to elevations of both external and internal ammonia levels. Branchial neuroepithelial cells (NECs) are thought to serve as internal sensors of plasma ammonia (peripheral chemoreceptors), but little is known about other possible ammonia-sensors. Here, we investigated whether trout possess external sensors and/or internal central chemoreceptors for ammonia. For external sensors, we analyzed the time course of ventilatory changes at the start of exposure to high environmental ammonia (HEA, 1 mM). Hyperventilation developed gradually over 20 min, suggesting that it was a response to internal ammonia elevation. We also directly perfused ammonia solutions (0.01-1 mM) to the external surfaces of the first gill arches. Immediate hypoventilation occurred. For central chemoreceptors, we injected ammonia solutions (0.5-1.0 mM) directly onto the surface of the hindbrain of anesthetized trout. Immediate hyperventilation occurred. This is the first evidence of central chemoreception in teleost fish. We conclude that trout possess both external ammonia sensors, and dual internal ammonia sensors (perhaps for redundancy), but their roles differ. External sensors cause short term hypoventilation, which would help limit toxic waterborne ammonia uptake. When fish cannot avoid HEA, the diffusion of waterborne ammonia into the blood will stimulate both peripheral (NECs) and central (brain) chemoreceptors, resulting in hyperventilation. This hyperventilation will be beneficial in increasing ammonia excretion via the Rh metabolon system in the gills not only after HEA exposure, but also after endogenous ammonia loading from feeding or exercise.


Assuntos
Amônia/sangue , Encéfalo/fisiologia , Brânquias/metabolismo , Oncorhynchus mykiss/fisiologia , Oxigênio/metabolismo , Amônia/química , Animais , Transporte Biológico , Encéfalo/metabolismo , Sistema Nervoso Central/fisiologia , Meio Ambiente , Hiperventilação , Células Neuroepiteliais/metabolismo , Fenômenos Fisiológicos Respiratórios , Água
20.
Artigo em Inglês | MEDLINE | ID: mdl-33059022

RESUMO

The effects of high external ammonia (HEA) exposure on breathing and the potential involvement of ammonia transporting Rh proteins in ammonia sensing were assessed in larval and adult zebrafish. Acute exposure of adults to either 250 or 500 µM (NH4)2SO4 caused increases in ventilation amplitude (AVENT) without affecting frequency (fVENT), resembling the ventilatory response to hypercapnia rather than hypoxia, during which fVENT was increased exclusively. The hyperventilatory response to HEA was prevented by hyperoxia, indicating that control of breathing through ammonia sensing is likely secondary to O2 chemoreception. Neuroepithelial cells (NECs) isolated from gill filaments exhibited a significant increase of intracellular [Ca2+] in response to 1 mM NH4Cl but this response was small (roughly 30%) compared to the response to hypercapnia (37.5 mmHg; ~800% increase). Immunohistochemistry (IHC) failed to reveal the presence of Rh proteins (Rhcgb, Rhbg or Rhag) in gill filament NECs. Knockout of rhcgb did not affect the ventilatory response of adults to HEA. Larvae at 4 days post fertilization (dpf) responded to HEA with increases in fVENT (AVENT was not measured). The hyperventilatory response of larvae to HEA was attenuated (60% reduction) after treatment from 0 to 4 dpf with the sympathetic neurotoxin 6-hydroxydopamine. In larvae, Rhcgb, Rhbg and Rhag were undetectable by IHC in cutaneous NECs yet the fVENT to HEA following Rhbg knockdown was slightly (22%) attenuated. Thus, the hyperventilatory response to external ammonia in adult zebrafish, while apparently initiated by activation of NECs, does not require Rhcgb, nor is the entry of ammonia into NECs reliant on other Rh proteins. The lack of colocalization of Rh proteins with NECs suggests that the entry of ammonia into NECs in larvae, also is not facilitated by this family of ammonia channels.


Assuntos
Amônia/farmacologia , Hiperventilação/fisiopatologia , Fenômenos Fisiológicos Respiratórios/efeitos dos fármacos , Peixe-Zebra/fisiologia , Amônia/metabolismo , Animais , Proteínas Sanguíneas/metabolismo , Cálcio/metabolismo , Proteínas de Transporte de Cátions/metabolismo , Brânquias/citologia , Brânquias/efeitos dos fármacos , Brânquias/metabolismo , Imuno-Histoquímica , Larva/citologia , Larva/efeitos dos fármacos , Larva/metabolismo , Glicoproteínas de Membrana/metabolismo , Células Neuroepiteliais/efeitos dos fármacos , Células Neuroepiteliais/metabolismo , Peixe-Zebra/genética , Peixe-Zebra/metabolismo , Proteínas de Peixe-Zebra/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA