Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
Add more filters










Publication year range
1.
Front Cell Dev Biol ; 11: 1268631, 2023.
Article in English | MEDLINE | ID: mdl-38020924

ABSTRACT

Tissue growth and morphogenesis are interrelated processes, whose tight coordination is essential for the production of different cell fates and the timely precise allocation of stem cell capacities. The zebrafish embryonic brainstem, the hindbrain, exemplifies such coupling between spatiotemporal cell diversity acquisition and tissue growth as the neurogenic commitment is differentially distributed over time. Here, we combined cell lineage and in vivo imaging approaches to reveal the emergence of specific cell population properties within the rhombomeres. We studied the molecular identity of hindbrain rhombomere centers and showed that they harbor different progenitor capacities that change over time. By clonal analysis, we revealed that cells within the center of rhombomeres decrease the proliferative capacity to remain mainly in the G1 phase. Proliferating progenitors give rise to neurons by asymmetric and symmetric neurogenic divisions while maintaining the pool of progenitors. The proliferative capacity of these cells differs from their neighbors, and they are delayed in the onset of Notch activity. Through functional studies, we demonstrated that they rely on Notch3 signaling to be maintained as non-committed progenitors. In this study, we show that cells in rhombomere centers, despite the neurogenic asynchrony, might share steps of a similar program with the rhombomere counterparts, to ensure proper tissue growth.

2.
Neuron ; 111(3): 345-361.e10, 2023 02 01.
Article in English | MEDLINE | ID: mdl-36417906

ABSTRACT

During development, regulatory factors appear in a precise order to determine cell fates over time. Consequently, to investigate complex tissue development, it is necessary to visualize and manipulate cell lineages with temporal control. Current strategies for tracing vertebrate cell lineages lack genetic access to sequentially produced cells. Here, we present TEMPO (Temporal Encoding and Manipulation in a Predefined Order), an imaging-readable genetic tool allowing differential labeling and manipulation of consecutive cell generations in vertebrates. TEMPO is based on CRISPR and powered by a cascade of gRNAs that drive orderly activation and inactivation of reporters and/or effectors. Using TEMPO to visualize zebrafish and mouse neurogenesis, we recapitulated birth-order-dependent neuronal fates. Temporally manipulating cell-cycle regulators in mouse cortex progenitors altered the proportion and distribution of neurons and glia, revealing the effects of temporal gene perturbation on serial cell fates. Thus, TEMPO enables sequential manipulation of molecular factors, crucial to study cell-type specification.


Subject(s)
Neurons , Zebrafish , Animals , Mice , Cell Lineage/physiology , Neurons/physiology , Neuroglia , Cell Differentiation/genetics , Neurogenesis/genetics , Gene Expression Regulation, Developmental
3.
Elife ; 112022 Sep 28.
Article in English | MEDLINE | ID: mdl-36169400

ABSTRACT

Reconstruction of prototypic three-dimensional (3D) atlases at the scale of whole tissues or organs requires specific methods to be developed. We have established a digital 3D-atlas maker (DAMAKER) and built a digital 3D-atlas to monitor the changes in the growth of the neuronal differentiation domain in the zebrafish hindbrain upon time. DAMAKER integrates spatial and temporal data of cell populations, neuronal differentiation and brain morphogenesis, through in vivo imaging techniques paired with image analyses and segmentation tools. First, we generated a 3D-reference from several imaged hindbrains and segmented them using a trainable tool; these were aligned using rigid registration, revealing distribution of neuronal differentiation growth patterns along the axes. Second, we quantified the dynamic growth of the neuronal differentiation domain by in vivo neuronal birthdating experiments. We generated digital neuronal birthdating 3D-maps and revealed that the temporal order of neuronal differentiation prefigured the spatial distribution of neurons in the tissue, with an inner-outer differentiation gradient. Last, we applied it to specific differentiated neuronal populations such as glutamatergic and GABAergic neurons, as proof-of-concept that the digital birthdating 3D-maps could be used as a proxy to infer neuronal birthdate. As this protocol uses open-access tools and algorithms, it can be shared for standardized, accessible, tissue-wide cell population atlas construction.


The brain, like most other organs, is formed by the coordinated growth of a few unspecialized cells in the embryo, which give rise to billions of neurons. For the brain to work properly, it is crucial that, during embryonic development, each neuron ends up in the correct location. This migration to the right spot has to happen while the brain grows and changes shape, which affects how and how far neurons and their precursor cells need to move to reach their final position. If these movements and changes in shape are not coordinated correctly, neurons can end up in the wrong place, form the wrong connections, and ultimately impact how the brain works. Previous work done in fruit flies and zebrafish resulted in three-dimensional maps of these animals' healthy brains, which allowed scientists to have a holistic view of how brains are organized. Although these maps are a valuable resource to study the structure of the brain, they do not provide information on how the brain transforms over time, especially during embryonic development. To get a clearer picture of how a few precursor cells give rise to the incredibly complex tissue that is the brain, a three-dimensional map spanning the entire developmental process is needed. To fill this gap in knowledge, Blanc et al. developed a digital atlas-maker pipeline (DAMAKER) that allows scientists to generate three-dimensional models of the embryonic brain from microscopy images of several individuals. They then used this pipeline to construct a three-dimensional digital atlas of how a part of the brain called the hindbrain develops in the zebrafish embryo. First, they collected images of the hindbrain showing neurons born at different times and matched these images to the existing static maps. Next, DAMAKER was used to follow neurons from the time of their birth to their final location, allowing Blanc et al. to create a map showing where neurons born at different stages during development end up. This type of map allows users to accurately determine when different populations of mature neurons are born, which allows scientists to estimate when different defects in brain development might originate. Based on these data, Blanc et al. concluded that in zebrafish most of the cells that will end up forming the hindbrain acquire their specialized neuronal identities very early in development, between 24 and 48 hours post fertilization. These temporal maps of healthy hindbrains were then compared to maps of brains in which the birth of neurons was disrupted, thus changing the final number of neurons in the brain. This experiment showed that changing the number of neurons that are born early in development alters the final positions of neurons and the overall shape of the brain. Therefore, for the brain to grow to its correct size, there must be a balance between the number of unspecialized cells in the developing brain, and the rate at which these cells become neurons. The DAMAKER pipeline not only provides scientists with a tool to study neurodevelopmental disorders, but also serves as a method that can be adjusted to map growth and shaping of other organs.


Subject(s)
Neurogenesis , Zebrafish , Animals , Rhombencephalon , Neurons , Image Processing, Computer-Assisted
4.
Cell Rep ; 39(10): 110915, 2022 06 07.
Article in English | MEDLINE | ID: mdl-35675784

ABSTRACT

Elucidating the cellular and molecular mechanisms that regulate the balance between progenitor cell proliferation and neuronal differentiation in the construction of the embryonic brain demands the combination of cell lineage and functional approaches. Here, we generate the comprehensive lineage of hindbrain boundary cells by using a CRISPR-based knockin zebrafish transgenic line that specifically labels the boundaries. We unveil that boundary cells asynchronously engage in neurogenesis undergoing a functional transition from neuroepithelial progenitors to radial glia cells, coinciding with the onset of Notch3 signaling that triggers their asymmetrical cell division. Upon notch3 loss of function, boundary cells lose radial glia properties and symmetrically divide undergoing neuronal differentiation. Finally, we show that the fate of boundary cells is to become neurons, the subtype of which relies on their axial position, suggesting that boundary cells contribute to refine the number and proportion of the distinct neuronal populations.


Subject(s)
Asymmetric Cell Division , Zebrafish , Animals , Cell Differentiation , Neurogenesis , Rhombencephalon/metabolism , Zebrafish/metabolism , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
5.
Sci Adv ; 7(44): eabi7785, 2021 Oct 29.
Article in English | MEDLINE | ID: mdl-34714681

ABSTRACT

Mechanical forces are exerted throughout cytokinesis, the final step of cell division. Yet, how forces are transduced and affect the signaling dynamics of cytokinetic proteins remains poorly characterized. We now show that the mechanosensitive Piezo1 channel is activated at the intercellular bridge (ICB) connecting daughter cells to regulate abscission. Inhibition of Piezo1 caused multinucleation both in vitro and in vivo. Piezo1 positioning at the ICB during cytokinesis depends on Pacsin3. Pharmacological and genetic inhibition of Piezo1 or Pacsin3 resulted in mislocation of Rab11-family-interacting protein 3 (Rab11-FIP3) endosomes, apoptosis-linked gene 2-interacting protein X (ALIX), and endosomal sorting complex required for transport III (ESCRT-III). Furthermore, we identified FIP3 as the link between Piezo1-generated Ca2+ signals and ALIX delivery to the ICB, where ALIX recruits the ESCRT-III component charged multivesicular body protein 4B, which promotes abscission. These results provide a different view of how mechanical forces participate in cytokinesis and identify Piezo1 as a key modulator of endosome trafficking.

6.
Front Cell Dev Biol ; 9: 711531, 2021.
Article in English | MEDLINE | ID: mdl-34490262

ABSTRACT

Cells in growing tissues receive both biochemical and physical cues from their microenvironment. Growing evidence has shown that mechanical signals are fundamental regulators of cell behavior. However, how physical properties of the microenvironment are transduced into critical cell behaviors, such as proliferation, progenitor maintenance, or differentiation during development, is still poorly understood. The transcriptional co-activators YAP/TAZ shuttle between the cytoplasm and the nucleus in response to multiple inputs and have emerged as important regulators of tissue growth and regeneration. YAP/TAZ sense and transduce physical cues, such as those from the extracellular matrix or the actomyosin cytoskeleton, to regulate gene expression, thus allowing them to function as gatekeepers of progenitor behavior in several developmental contexts. The Notch pathway is a key signaling pathway that controls binary cell fate decisions through cell-cell communication in a context-dependent manner. Recent reports now suggest that the crosstalk between these two pathways is critical for maintaining the balance between progenitor maintenance and cell differentiation in different tissues. How this crosstalk integrates with morphogenesis and changes in tissue architecture during development is still an open question. Here, we discuss how progenitor cell proliferation, specification, and differentiation are coordinated with morphogenesis to construct a functional organ. We will pay special attention to the interplay between YAP/TAZ and Notch signaling pathways in determining cell fate decisions and discuss whether this represents a general mechanism of regulating cell fate during development. We will focus on research carried out in vertebrate embryos that demonstrate the important roles of mechanical cues in stem cell biology and discuss future challenges.

7.
Front Neurosci ; 15: 781160, 2021.
Article in English | MEDLINE | ID: mdl-35046768

ABSTRACT

The central nervous system (CNS) exhibits an extraordinary diversity of neurons, with the right cell types and proportions at the appropriate sites. Thus, to produce brains with specific size and cell composition, the rates of proliferation and differentiation must be tightly coordinated and balanced during development. Early on, proliferation dominates; later on, the growth rate almost ceases as more cells differentiate and exit the cell cycle. Generation of cell diversity and morphogenesis takes place concomitantly. In the vertebrate brain, this results in dramatic changes in the position of progenitor cells and their neuronal derivatives, whereas in the spinal cord morphogenetic changes are not so important because the structure mainly grows by increasing its volume. Morphogenesis is under control of specific genetic programs that coordinately unfold over time; however, little is known about how they operate and impact in the pools of progenitor cells in the CNS. Thus, the spatiotemporal coordination of these processes is fundamental for generating functional neuronal networks. Some key aims in developmental neurobiology are to determine how cell diversity arises from pluripotent progenitor cells, and how the progenitor potential changes upon time. In this review, we will share our view on how the advance of new technologies provides novel data that challenge some of the current hypothesis. We will cover some of the latest studies on cell lineage tracing and clonal analyses addressing the role of distinct progenitor cell division modes in balancing the rate of proliferation and differentiation during brain morphogenesis. We will discuss different hypothesis proposed to explain how progenitor cell diversity is generated and how they challenged prevailing concepts and raised new questions.

8.
Semin Cell Dev Biol ; 107: 179-189, 2020 11.
Article in English | MEDLINE | ID: mdl-32448645

ABSTRACT

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.


Subject(s)
Rhombencephalon/cytology , Rhombencephalon/embryology , Animals , Cell Proliferation , Embryonic Development , Humans , Mechanotransduction, Cellular , Models, Biological , Phylogeny
9.
PLoS One ; 15(2): e0228225, 2020.
Article in English | MEDLINE | ID: mdl-32012186

ABSTRACT

The Lower Rhombic Lip (LRL) is a transient neuroepithelial structure of the dorsal hindbrain, which expands from r2 to r7, and gives rise to deep nuclei of the brainstem, such as the vestibular and auditory nuclei and most posteriorly the precerebellar nuclei. Although there is information about the contribution of specific proneural-progenitor populations to specific deep nuclei, and the distinct rhombomeric contribution, little is known about how progenitor cells from the LRL behave during neurogenesis and how their transition into differentiation is regulated. In this work, we investigated the atoh1 gene regulatory network operating in the specification of LRL cells, and the kinetics of cell proliferation and behavior of atoh1a-derivatives by using complementary strategies in the zebrafish embryo. We unveiled that atoh1a is necessary and sufficient for specification of LRL cells by activating atoh1b, which worked as a differentiation gene to transition progenitor cells towards neuron differentiation in a Notch-dependent manner. This cell state transition involved the release of atoh1a-derivatives from the LRL: atoh1a progenitors contributed first to atoh1b cells, which are committed non-proliferative precursors, and to the lhx2b-neuronal lineage as demonstrated by cell fate studies and functional analyses. Using in vivo cell lineage approaches we revealed that the proliferative cell capacity, as well as the mode of division, relied on the position of the atoh1a progenitors within the dorsoventral axis. We showed that atoh1a may behave as the cell fate selector gene, whereas atoh1b functions as a neuronal differentiation gene, contributing to the lhx2b neuronal population. atoh1a-progenitor cell dynamics (cell proliferation, cell differentiation, and neuronal migration) relies on their position, demonstrating the challenges that progenitor cells face in computing positional information from a dynamic two-dimensional grid in order to generate the stereotyped neuronal structures in the embryonic hindbrain.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Metencephalon/metabolism , Morphogenesis/genetics , Rhombencephalon/growth & development , Transcription Factors/genetics , Zebrafish Proteins/genetics , Animals , Gene Expression Regulation, Developmental , Imaging, Three-Dimensional , Neurons/cytology , Rhombencephalon/cytology , Zebrafish/genetics , Zebrafish/growth & development
10.
Development ; 146(14)2019 07 22.
Article in English | MEDLINE | ID: mdl-31273051

ABSTRACT

Cells perceive their microenvironment through chemical and physical cues. However, how the mechanical signals are interpreted during embryonic tissue deformation to result in specific cell behaviors is largely unknown. The Yap/Taz family of transcriptional co-activators has emerged as an important regulator of tissue growth and regeneration, responding to physical cues from the extracellular matrix, and to cell shape and actomyosin cytoskeletal changes. In this study, we demonstrate the role of Yap/Taz-TEAD activity as a sensor of mechanical signals in the regulation of the progenitor behavior of boundary cells during zebrafish hindbrain compartmentalization. Monitoring of in vivo Yap/Taz activity during hindbrain segmentation indicated that boundary cells responded to mechanical cues in a cell-autonomous manner through Yap/Taz-TEAD activity. Cell-lineage analysis revealed that Yap/Taz-TEAD boundary cells decreased their proliferative activity when Yap/Taz-TEAD activity ceased, which preceded changes in their cell fate from proliferating progenitors to differentiated neurons. Functional experiments demonstrated the pivotal role of Yap/Taz-TEAD signaling in maintaining progenitor features in the hindbrain boundary cell population.


Subject(s)
Cell Division/genetics , DNA-Binding Proteins/physiology , Intracellular Signaling Peptides and Proteins/physiology , Nuclear Proteins/physiology , Rhombencephalon/cytology , Rhombencephalon/embryology , Stem Cells/physiology , Trans-Activators/physiology , Transcription Factors/physiology , Zebrafish Proteins/physiology , Animals , Animals, Genetically Modified , Body Patterning/genetics , Cell Differentiation/genetics , Cell Movement/genetics , DNA-Binding Proteins/genetics , Embryo, Nonmammalian , Intracellular Signaling Peptides and Proteins/genetics , Mechanical Phenomena , Mechanotransduction, Cellular/genetics , Mechanotransduction, Cellular/physiology , Neurogenesis/genetics , Nuclear Proteins/genetics , Organogenesis/genetics , Rhombencephalon/metabolism , Signal Transduction/genetics , Stem Cells/cytology , TEA Domain Transcription Factors , Trans-Activators/genetics , Transcription Factors/genetics , Transcriptional Coactivator with PDZ-Binding Motif Proteins , YAP-Signaling Proteins , Zebrafish/embryology , Zebrafish/genetics , Zebrafish Proteins/genetics
11.
J Clin Invest ; 129(3): 1240-1256, 2019 03 01.
Article in English | MEDLINE | ID: mdl-30620337

ABSTRACT

Sphingolipid imbalance is the culprit in a variety of neurological diseases, some affecting the myelin sheath. We have used whole-exome sequencing in patients with undetermined leukoencephalopathies to uncover the endoplasmic reticulum lipid desaturase DEGS1 as the causative gene in 19 patients from 13 unrelated families. Shared features among the cases include severe motor arrest, early nystagmus, dystonia, spasticity, and profound failure to thrive. MRI showed hypomyelination, thinning of the corpus callosum, and progressive thalamic and cerebellar atrophy, suggesting a critical role of DEGS1 in myelin development and maintenance. This enzyme converts dihydroceramide (DhCer) into ceramide (Cer) in the final step of the de novo biosynthesis pathway. We detected a marked increase of the substrate DhCer and DhCer/Cer ratios in patients' fibroblasts and muscle. Further, we used a knockdown approach for disease modeling in Danio rerio, followed by a preclinical test with the first-line treatment for multiple sclerosis, fingolimod (FTY720, Gilenya). The enzymatic inhibition of Cer synthase by fingolimod, 1 step prior to DEGS1 in the pathway, reduced the critical DhCer/Cer imbalance and the severe locomotor disability, increasing the number of myelinating oligodendrocytes in a zebrafish model. These proof-of-concept results pave the way to clinical translation.


Subject(s)
Animals, Genetically Modified , Brain , Fingolimod Hydrochloride/pharmacology , Hereditary Central Nervous System Demyelinating Diseases , Zebrafish Proteins , Zebrafish , Animals , Animals, Genetically Modified/genetics , Animals, Genetically Modified/metabolism , Brain/enzymology , Brain/pathology , Disease Models, Animal , Fatty Acid Desaturases/genetics , Fatty Acid Desaturases/metabolism , Hereditary Central Nervous System Demyelinating Diseases/drug therapy , Hereditary Central Nervous System Demyelinating Diseases/enzymology , Hereditary Central Nervous System Demyelinating Diseases/genetics , Hereditary Central Nervous System Demyelinating Diseases/pathology , Humans , Locomotion/drug effects , Oligodendroglia/enzymology , Oligodendroglia/pathology , Zebrafish/genetics , Zebrafish/metabolism , Zebrafish Proteins/genetics
12.
Proc Natl Acad Sci U S A ; 115(16): E3731-E3740, 2018 04 17.
Article in English | MEDLINE | ID: mdl-29610331

ABSTRACT

Developmental programs often rely on parallel morphogenetic mechanisms that guarantee precise tissue architecture. While redundancy constitutes an obvious selective advantage, little is known on how novel morphogenetic mechanisms emerge during evolution. In zebrafish, rhombomeric boundaries behave as an elastic barrier, preventing cell intermingling between adjacent compartments. Here, we identify the fundamental role of the small-GTPase Rac3b in actomyosin cable assembly at hindbrain boundaries. We show that the novel rac3b/rfng/sgca regulatory cluster, which is specifically expressed at the boundaries, emerged in the Ostariophysi superorder by chromosomal rearrangement that generated new cis-regulatory interactions. By combining 4C-seq, ATAC-seq, transgenesis, and CRISPR-induced deletions, we characterized this regulatory domain, identifying hindbrain boundary-specific cis-regulatory elements. Our results suggest that the capacity of boundaries to act as an elastic mesh for segregating rhombomeric cells evolved by cooption of critical genes to a novel regulatory block, refining the mechanisms for hindbrain segmentation.


Subject(s)
Actomyosin/physiology , Gene Expression Regulation, Developmental , Rhombencephalon/embryology , Sarcoglycans/physiology , Zebrafish Proteins/physiology , Zebrafish/embryology , rac GTP-Binding Proteins/physiology , Animals , Body Patterning/genetics , CRISPR-Cas Systems , Cell Movement , Characidae/genetics , Characidae/physiology , Chromatin/genetics , Chromatin/ultrastructure , Evolution, Molecular , Fishes/classification , Fishes/genetics , Morphogenesis , Mutagenesis, Site-Directed , Neurogenesis , Phylogeny , Sarcoglycans/genetics , Species Specificity , Zebrafish/genetics , Zebrafish Proteins/genetics , rac GTP-Binding Proteins/genetics
13.
Elife ; 62017 01 04.
Article in English | MEDLINE | ID: mdl-28051766

ABSTRACT

Reconstructing the lineage of cells is central to understanding how the wide diversity of cell types develops. Here, we provide the neurosensory lineage reconstruction of a complex sensory organ, the inner ear, by imaging zebrafish embryos in vivo over an extended timespan, combining cell tracing and cell fate marker expression over time. We deliver the first dynamic map of early neuronal and sensory progenitor pools in the whole otic vesicle. It highlights the remodeling of the neuronal progenitor domain upon neuroblast delamination, and reveals that the order and place of neuroblasts' delamination from the otic epithelium prefigure their position within the SAG. Sensory and non-sensory domains harbor different proliferative activity contributing distinctly to the overall growth of the structure. Therefore, the otic vesicle case exemplifies a generic morphogenetic process where spatial and temporal cues regulate cell fate and functional organization of the rudiment of the definitive organ.


Subject(s)
Cell Lineage , Ear, Inner/cytology , Ear, Inner/embryology , Morphogenesis , Sensory Receptor Cells/physiology , Stem Cells/physiology , Zebrafish , Animals , Optical Imaging
14.
Genesis ; 53(9): 583-603, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26174808

ABSTRACT

Angiogenesis is an essential requirement for embryonic development and adult homeostasis. Its deregulation is a key feature of numerous pathologies and many studies have shown that members of the transforming growth factor beta (TGF-ß) family of proteins play important roles in angiogenesis during development and disease. Betaglycan (BG), also known as TGF-ß receptor type III, is a TGF-ß coreceptor essential for mice embryonic development but its role in angiogenesis has not been described. We have cloned the cDNA encoding zebrafish BG, a TGF-ß-binding membrane proteoglycan that showed a dynamic expression pattern in zebrafish embryos, including the notochord and cells adjacent to developing vessels. Injection of antisense morpholinos decreased BG protein levels and morphant embryos exhibited impaired angiogenesis that was rescued by coinjection with rat BG mRNA. In vivo time-lapse microscopy revealed that BG deficiency differentially affected arterial and venous angiogenesis: morphants showed impaired pathfinding of intersegmental vessels migrating from dorsal aorta, while endothelial cells originating from the caudal vein displayed sprouting and migration defects. Our results reveal a new role for BG during embryonic angiogenesis in zebrafish, which has not been described in mammals and pose interesting questions about the molecular machinery regulating angiogenesis in different vertebrates. genesis 53:583-603, 2015. © 2015 Wiley Periodicals, Inc.

15.
Cell Mol Life Sci ; 72(19): 3721-30, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26089248

ABSTRACT

Segregating cells into compartments during embryonic development is essential for growth and pattern formation. In the developing hindbrain, boundaries separate molecularly, physically and neuroanatomically distinct segments called rhombomeres. After rhombomeric cells have acquired their identity, interhombomeric boundaries restrict cell intermingling between adjacent rhombomeres and act as signaling centers to pattern the surrounding tissue. Several works have stressed the relevance of Eph/ephrin signaling in rhombomeric cell sorting. Recent data have unveiled the role of this pathway in the assembly of actomyosin cables as an important mechanism for keeping cells from different rhombomeres segregated. In this Review, we will provide a short summary of recent evidences gathered in different systems suggesting that physical actomyosin barriers can be a general mechanism for tissue separation. We will discuss current evidences supporting a model where cell-cell signaling pathways, such as Eph/ephrin, govern compartmental cell sorting through modulation of the actomyosin cytoskeleton and cell adhesive properties to prevent cell intermingling.


Subject(s)
Cell Communication/physiology , Cell Differentiation/physiology , Cell Movement/physiology , Models, Neurological , Morphogenesis/physiology , Rhombencephalon/embryology , Actomyosin/biosynthesis , Animals , Cell Adhesion/physiology , Humans , Species Specificity
16.
J Neurosci ; 35(19): 7475-86, 2015 May 13.
Article in English | MEDLINE | ID: mdl-25972174

ABSTRACT

Establishing topographical maps of the external world is an important but still poorly understood feature of the vertebrate sensory system. To study the selective innervation of hindbrain regions by sensory afferents in the zebrafish embryo, we mapped the fine-grained topographical representation of sensory projections at the central level by specific photoconversion of sensory neurons. Sensory ganglia located anteriorly project more medially than do ganglia located posteriorly, and this relates to the order of sensory ganglion differentiation. By single-plane illumination microscopy (SPIM) in vivo imaging, we show that (1) the sequence of arrival of cranial ganglion inputs predicts the topography of central projections, and (2) delaminated neuroblasts differentiate in close contact with the neural tube, and they never loose contact with the neural ectoderm. Afferent entrance points are established by plasma membrane interactions between primary differentiated peripheral sensory neurons and neural tube border cells with the cooperation of neural crest cells. These first contacts remain during ensuing morphological growth to establish pioneer axons. Neural crest cells and repulsive slit1/robo2 signals then guide axons from later-differentiating neurons toward the neural tube. Thus, this study proposes a new model by which the topographical representation of cranial sensory ganglia is established by entrance order, with the entry points determined by cell contact between the sensory ganglion cell bodies and the hindbrain.


Subject(s)
Afferent Pathways/physiology , Brain Mapping , Cell Differentiation/physiology , Gene Expression Regulation, Developmental/physiology , Rhombencephalon/anatomy & histology , Sensory Receptor Cells/physiology , Afferent Pathways/drug effects , Animals , Animals, Genetically Modified , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Differentiation/genetics , Chemokine CXCL12/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Embryo, Nonmammalian , Enzyme Inhibitors/pharmacology , Female , Gene Expression Regulation, Developmental/genetics , Isoxazoles/pharmacology , LIM-Homeodomain Proteins/genetics , LIM-Homeodomain Proteins/metabolism , Leflunomide , Male , Morpholinos/pharmacology , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neural Tube/cytology , Rhombencephalon/drug effects , Rhombencephalon/embryology , Sensory Receptor Cells/drug effects , Signal Transduction/genetics , Signal Transduction/physiology , Transcription Factors/genetics , Transcription Factors/metabolism , Zebrafish , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
17.
Birth Defects Res A Clin Mol Teratol ; 103(3): 163-77, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25131946

ABSTRACT

Ethanol is the most common human teratogen, and its consumption during pregnancy can produce a wide range of abnormalities in infants known as fetal alcohol spectrum disorder (FASD). The major characteristics of FASD can be divided into: (i) growth retardation, (ii) craniofacial abnormalities, and (iii) central nervous system (CNS) dysfunction. FASD is the most common cause of nongenetic mental retardation in Western countries. Although the underlying molecular mechanisms of ethanol neurotoxicity are not completely determined, the induction of oxidative stress is believed to be one central process linked to the development of the disease. Currently, there is no known effective strategy for prevention (other than alcohol avoidance) or treatment. In the present review we will provide the state of art in the evidence for the use of antioxidants as a potential therapeutic strategy for the treatment using whole-embryo and culture cells models of FASD. We conclude that the imbalance of the intracellular redox state contributes to the pathogenesis observed in FASD models, and we suggest that antioxidant therapy can be considered a new efficient strategy to mitigate the effects of prenatal ethanol exposure.


Subject(s)
Antioxidants/therapeutic use , Fetal Alcohol Spectrum Disorders/prevention & control , Ethanol/toxicity , Female , Humans , Pregnancy , Teratogens/toxicity
18.
PLoS One ; 9(11): e112851, 2014.
Article in English | MEDLINE | ID: mdl-25383948

ABSTRACT

BACKGROUND: The exposure of the human embryo to ethanol results in a spectrum of disorders involving multiple organ systems, including the impairment of the development of the central nervous system (CNS). In spite of the importance for human health, the molecular basis of prenatal ethanol exposure remains poorly understood, mainly to the difficulty of sample collection. Zebrafish is now emerging as a powerful organism for the modeling and the study of human diseases. In this work, we have assessed the sensitivity of specific subsets of neurons to ethanol exposure during embryogenesis and we have visualized the sensitive embryonic developmental periods for specific neuronal groups by the use of different transgenic zebrafish lines. METHODOLOGY/PRINCIPAL FINDINGS: In order to evaluate the teratogenic effects of acute ethanol exposure, we exposed zebrafish embryos to ethanol in a given time window and analyzed the effects in neurogenesis, neuronal differentiation and brain patterning. Zebrafish larvae exposed to ethanol displayed small eyes and/or a reduction of the body length, phenotypical features similar to the observed in children with prenatal exposure to ethanol. When neuronal populations were analyzed, we observed a clear reduction in the number of differentiated neurons in the spinal cord upon ethanol exposure. There was a decrease in the population of sensory neurons mainly due to a decrease in cell proliferation and subsequent apoptosis during neuronal differentiation, with no effect in motoneuron specification. CONCLUSION: Our investigation highlights that transient exposure to ethanol during early embryonic development affects neuronal differentiation although does not result in defects in early neurogenesis. These results establish the use of zebrafish embryos as an alternative research model to elucidate the molecular mechanism(s) of ethanol-induced developmental toxicity at very early stages of embryonic development.


Subject(s)
Embryo, Nonmammalian/abnormalities , Embryonic Development/drug effects , Ethanol/toxicity , Neurons/drug effects , Animals , Body Patterning/drug effects , Cell Differentiation/drug effects , Disease Models, Animal , Embryo, Nonmammalian/drug effects , Fetal Alcohol Spectrum Disorders/physiopathology , Gene Expression Regulation, Developmental/drug effects , Humans , Neurogenesis/drug effects , Zebrafish
19.
EMBO J ; 33(7): 686-701, 2014 Apr 01.
Article in English | MEDLINE | ID: mdl-24569501

ABSTRACT

Segregating cells into compartments during embryonic development is essential for growth and pattern formation. Physical mechanisms shaping compartment boundaries were recently explored in Drosophila, where actomyosin-based barriers were revealed to be important for keeping cells apart. In vertebrates, interhombomeric boundaries are straight interfaces, which often serve as signaling centers that pattern the surrounding tissue. Here, we demonstrate that in the hindbrain of zebrafish embryos cell sorting sharpens the molecular boundaries and, once borders are straight, actomyosin barriers are key to keeping rhombomeric cells segregated. Actomyosin cytoskeletal components are enriched at interhombomeric boundaries, forming cable-like structures in the apical side of the neuroepithelial cells by the time morphological boundaries are visible. When myosin II function is inhibited, cable structures do not form, leading to rhombomeric cell mixing. Downregulation of EphA4a compromises actomyosin cables and cells with different rhombomeric identity intermingle, and the phenotype is rescued enhancing myosin II activity. Moreover, enrichment of actomyosin structures is obtained when EphA4 is ectopically expressed in even-numbered rhombomeres. These findings suggest that mechanical barriers act downstream of EphA/ephrin signaling to segregate cells from different rhombomeres.


Subject(s)
Actomyosin/metabolism , Cytoskeleton/metabolism , Gene Expression Regulation, Developmental , Receptor, EphA4/metabolism , Rhombencephalon/embryology , Zebrafish/embryology , Animals , Cell Division , Cell Movement , Down-Regulation , Embryonic Development/physiology , Ephrins/metabolism , Female , Genes, Reporter , Myosin Type II/metabolism , Organisms, Genetically Modified , Rhombencephalon/metabolism , Rhombencephalon/ultrastructure , Signal Transduction , Zebrafish/metabolism
20.
Mech Dev ; 130(2-3): 160-8, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23041177

ABSTRACT

Fgf and Wnt signalling have been shown to be required for formation of the otic placode in vertebrates. Whereas several Fgfs including Fgf3, Fgf8 and Fgf10 have been shown to participate during early placode induction, Wnt signalling is required for specification and maintenance of the otic placode, and dorsal patterning of the otic vesicle. However, the requirement for specific members of the Wnt gene family for otic placode and vesicle formation and their potential interaction with Fgf signalling has been poorly defined. Due to its spatiotemporal expression during placode formation in the hindbrain Wnt8a has been postulated as a potential candidate for its specification. Here we have examined the role of Wnt8a during formation of the otic placode and vesicle in mouse embryos. Wnt8a expression depends on the presence of Fgf3 indicating a serial regulation between Fgf and Wnt signalling during otic placode induction and specification. Wnt8a by itself however is neither essential for placode specification nor redundantly required together with Fgfs for otic placode and vesicle formation. Interestingly however, Wnt8a and Fgf3 are redundantly required for expression of Fgf15 in the hindbrain indicating additional reciprocal interactions between Fgf and Wnt signalling. Further reduction of Wnt signalling by the inactivation of Wnt1 in a Wnt8a mutant background revealed a redundant requirement for both genes during morphogenesis of the dorsal portion of the otic vesicle.


Subject(s)
Body Patterning/genetics , Endolymphatic Duct/embryology , Intercellular Signaling Peptides and Proteins/metabolism , Animals , Down-Regulation , Ear, Inner/embryology , Ear, Inner/metabolism , Endolymphatic Duct/metabolism , Fibroblast Growth Factor 3/genetics , Fibroblast Growth Factor 3/metabolism , Fibroblast Growth Factors/genetics , Fibroblast Growth Factors/metabolism , Gene Deletion , Gene Expression , Gene Expression Regulation, Developmental , Intercellular Signaling Peptides and Proteins/genetics , Mice , Mice, Inbred C57BL , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Rhombencephalon/embryology , Rhombencephalon/metabolism , Wnt Proteins/genetics , Wnt Proteins/metabolism , Wnt Signaling Pathway , Wnt1 Protein/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...