Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 33
2.
Nat Commun ; 14(1): 3060, 2023 05 27.
Article En | MEDLINE | ID: mdl-37244931

Formation of oriented myofibrils is a key event in musculoskeletal development. However, the mechanisms that drive myocyte orientation and fusion to control muscle directionality in adults remain enigmatic. Here, we demonstrate that the developing skeleton instructs the directional outgrowth of skeletal muscle and other soft tissues during limb and facial morphogenesis in zebrafish and mouse. Time-lapse live imaging reveals that during early craniofacial development, myoblasts condense into round clusters corresponding to future muscle groups. These clusters undergo oriented stretch and alignment during embryonic growth. Genetic perturbation of cartilage patterning or size disrupts the directionality and number of myofibrils in vivo. Laser ablation of musculoskeletal attachment points reveals tension imposed by cartilage expansion on the forming myofibers. Application of continuous tension using artificial attachment points, or stretchable membrane substrates, is sufficient to drive polarization of myocyte populations in vitro. Overall, this work outlines a biomechanical guidance mechanism that is potentially useful for engineering functional skeletal muscle.


Muscle, Skeletal , Zebrafish , Animals , Mice , Zebrafish/genetics , Muscle, Skeletal/physiology , Myofibrils/physiology , Morphogenesis , Myoblasts/physiology
3.
Philos Trans R Soc Lond B Biol Sci ; 378(1880): 20220079, 2023 07 03.
Article En | MEDLINE | ID: mdl-37183897

Extensive morphological variation found in mammals reflects the wide spectrum of their ecological adaptations. The highest morphological diversity is present in the craniofacial region, where geometry is mainly dictated by the bony skull. Mammalian craniofacial development represents complex multistep processes governed by numerous conserved genes that require precise spatio-temporal control. A central question in contemporary evolutionary biology is how a defined set of conserved genes can orchestrate formation of fundamentally different structures, and therefore how morphological variability arises. In principle, differential gene expression patterns during development are the source of morphological variation. With the emergence of multicellular organisms, precise regulation of gene expression in time and space is attributed to cis-regulatory elements. These elements contribute to higher-order chromatin structure and together with trans-acting factors control transcriptional landscapes that underlie intricate morphogenetic processes. Consequently, divergence in cis-regulation is believed to rewire existing gene regulatory networks and form the core of morphological evolution. This review outlines the fundamental principles of the genetic code and genomic regulation interplay during development. Recent work that deepened our comprehension of cis-regulatory element origin, divergence and function is presented here to illustrate the state-of-the-art research that uncovered the principles of morphological novelty. This article is part of the theme issue 'The mammalian skull: development, structure and function'.


Evolution, Molecular , Mammals , Animals , Mammals/genetics , Gene Regulatory Networks , Skull , Head
4.
Philos Trans R Soc Lond B Biol Sci ; 378(1880): 20220080, 2023 07 03.
Article En | MEDLINE | ID: mdl-37183900

Mammals possess impressive craniofacial variation that mirrors their adaptation to diverse ecological niches, feeding behaviour, physiology and overall lifestyle. The spectrum of craniofacial geometries is established mainly during embryonic development. The formation of the head represents a sequence of events regulated on genomic, molecular, cellular and tissue level, with each step taking place under tight spatio-temporal control. Even minor variations in timing, position or concentration of the molecular drivers and the resulting events can affect the final shape, size and position of the skeletal elements and the geometry of the head. Our knowledge of craniofacial development increased substantially in the last decades, mainly due to research using conventional vertebrate model organisms. However, how developmental differences in head formation arise specifically within mammals remains largely unexplored. This review highlights three evolutionary mechanisms acknowledged to modify ontogenesis: heterochrony, heterotopy and heterometry. We present recent research that links changes in developmental timing, spatial organization or gene expression levels to the acquisition of species-specific skull morphologies. We highlight how these evolutionary modifications occur on the level of the genes, molecules and cellular processes, and alter conserved developmental programmes to generate a broad spectrum of skull shapes characteristic of the class Mammalia. This article is part of the theme issue 'The mammalian skull: development, structure and function'.


Head , Skull , Animals , Biological Evolution , Mammals , Species Specificity
5.
Nat Commun ; 14(1): 3092, 2023 05 29.
Article En | MEDLINE | ID: mdl-37248239

In this study we use comparative genomics to uncover a gene with uncharacterized function (1700011H14Rik/C14orf105/CCDC198), which we hereby name FAME (Factor Associated with Metabolism and Energy). We observe that FAME shows an unusually high evolutionary divergence in birds and mammals. Through the comparison of single nucleotide polymorphisms, we identify gene flow of FAME from Neandertals into modern humans. We conduct knockout experiments on animals and observe altered body weight and decreased energy expenditure in Fame knockout animals, corresponding to genome-wide association studies linking FAME with higher body mass index in humans. Gene expression and subcellular localization analyses reveal that FAME is a membrane-bound protein enriched in the kidneys. Although the gene knockout results in structurally normal kidneys, we detect higher albumin in urine and lowered ferritin in the blood. Through experimental validation, we confirm interactions between FAME and ferritin and show co-localization in vesicular and plasma membranes.


Energy Metabolism , Genome-Wide Association Study , Animals , Humans , Body Weight , Energy Metabolism/genetics , Ferritins/genetics , Kidney , Neanderthals
6.
Nat Commun ; 13(1): 6949, 2022 11 14.
Article En | MEDLINE | ID: mdl-36376278

There are major differences in duration and scale at which limb development and regeneration proceed, raising the question to what extent regeneration is a recapitulation of development. We address this by analyzing skeletal elements using a combination of micro-CT imaging, molecular profiling and clonal cell tracing. We find that, in contrast to development, regenerative skeletal growth is accomplished based entirely on cartilage expansion prior to ossification, not limiting the transversal cartilage expansion and resulting in bulkier skeletal parts. The oriented extension of salamander cartilage and bone appear similar to the development of basicranial synchondroses in mammals, as we found no evidence for cartilage stem cell niches or growth plate-like structures during neither development nor regeneration. Both regenerative and developmental ossification in salamanders start from the cortical bone and proceeds inwards, showing the diversity of schemes for the synchrony of cortical and endochondral ossification among vertebrates.


Osteogenesis , Urodela , Animals , Bone and Bones , Cartilage , Cell Division , Mammals
7.
EMBO J ; 41(17): e108780, 2022 09 01.
Article En | MEDLINE | ID: mdl-35815410

Schwann cell precursors (SCPs) are nerve-associated progenitors that can generate myelinating and non-myelinating Schwann cells but also are multipotent like the neural crest cells from which they originate. SCPs are omnipresent along outgrowing peripheral nerves throughout the body of vertebrate embryos. By using single-cell transcriptomics to generate a gene expression atlas of the entire neural crest lineage, we show that early SCPs and late migratory crest cells have similar transcriptional profiles characterised by a multipotent "hub" state containing cells biased towards traditional neural crest fates. SCPs keep diverging from the neural crest after being primed towards terminal Schwann cells and other fates, with different subtypes residing in distinct anatomical locations. Functional experiments using CRISPR-Cas9 loss-of-function further show that knockout of the common "hub" gene Sox8 causes defects in neural crest-derived cells along peripheral nerves by facilitating differentiation of SCPs towards sympathoadrenal fates. Finally, specific tumour populations found in melanoma, neurofibroma and neuroblastoma map to different stages of SCP/Schwann cell development. Overall, SCPs resemble migrating neural crest cells that maintain multipotency and become transcriptionally primed towards distinct lineages.


Neural Crest , Schwann Cells , Cell Differentiation/physiology , Neurogenesis/physiology , Peripheral Nerves , Schwann Cells/metabolism
8.
Sci Rep ; 12(1): 8728, 2022 05 24.
Article En | MEDLINE | ID: mdl-35610276

The complex shape of embryonic cartilage represents a true challenge for phenotyping and basic understanding of skeletal development. X-ray computed microtomography (µCT) enables inspecting relevant tissues in all three dimensions; however, most 3D models are still created by manual segmentation, which is a time-consuming and tedious task. In this work, we utilised a convolutional neural network (CNN) to automatically segment the most complex cartilaginous system represented by the developing nasal capsule. The main challenges of this task stem from the large size of the image data (over a thousand pixels in each dimension) and a relatively small training database, including genetically modified mouse embryos, where the phenotype of the analysed structures differs from the norm. We propose a CNN-based segmentation model optimised for the large image size that we trained using a unique manually annotated database. The segmentation model was able to segment the cartilaginous nasal capsule with a median accuracy of 84.44% (Dice coefficient). The time necessary for segmentation of new samples shortened from approximately 8 h needed for manual segmentation to mere 130 s per sample. This will greatly accelerate the throughput of µCT analysis of cartilaginous skeletal elements in animal models of developmental diseases.


Deep Learning , Animals , Cartilage/diagnostic imaging , Developmental Biology , Image Processing, Computer-Assisted/methods , Mice , Neural Networks, Computer , X-Rays
9.
Gigascience ; 112022 04 05.
Article En | MEDLINE | ID: mdl-35380661

BACKGROUND: Lightless caves can harbour a wide range of living organisms. Cave animals have evolved a set of morphological, physiological, and behavioural adaptations known as troglomorphisms, enabling their survival in the perpetual darkness, narrow temperature and humidity ranges, and nutrient scarcity of the subterranean environment. In this study, we focused on adaptations of skull shape and sensory systems in the blind cave salamander, Proteus anguinus, also known as olm or simply proteus-the largest cave tetrapod and the only European amphibian living exclusively in subterranean environments. This extraordinary amphibian compensates for the loss of sight by enhanced non-visual sensory systems including mechanoreceptors, electroreceptors, and chemoreceptors. We compared developmental stages of P. anguinus with Ambystoma mexicanum, also known as axolotl, to make an exemplary comparison between cave- and surface-dwelling paedomorphic salamanders. FINDINGS: We used contrast-enhanced X-ray computed microtomography for the 3D segmentation of the soft tissues in the head of P. anguinus and A. mexicanum. Sensory organs were visualized to elucidate how the animal is adapted to living in complete darkness. X-ray microCT datasets were provided along with 3D models for larval, juvenile, and adult specimens, showing the cartilage of the chondrocranium and the position, shape, and size of the brain, eyes, and olfactory epithelium. CONCLUSIONS: P. anguinus still keeps some of its secrets. Our high-resolution X-ray microCT scans together with 3D models of the anatomical structures in the head may help to elucidate the nature and origin of the mechanisms behind its adaptations to the subterranean environment, which led to a series of troglomorphisms.


Proteidae , Animals , Darkness , Urodela , X-Rays
10.
Cell Mol Life Sci ; 78(16): 6033-6049, 2021 Aug.
Article En | MEDLINE | ID: mdl-34274976

Melanocytes are pigmented cells residing mostly in the skin and hair follicles of vertebrates, where they contribute to colouration and protection against UV-B radiation. However, the spectrum of their functions reaches far beyond that. For instance, these pigment-producing cells are found inside the inner ear, where they contribute to the hearing function, and in the heart, where they are involved in the electrical conductivity and support the stiffness of cardiac valves. The embryonic origin of such extracutaneous melanocytes is not clear. We took advantage of lineage-tracing experiments combined with 3D visualizations and gene knockout strategies to address this long-standing question. We revealed that Schwann cell precursors are recruited from the local innervation during embryonic development and give rise to extracutaneous melanocytes in the heart, brain meninges, inner ear, and other locations. In embryos with a knockout of the EdnrB receptor, a condition imitating Waardenburg syndrome, we observed only nerve-associated melanoblasts, which failed to detach from the nerves and to enter the inner ear. Finally, we looked into the evolutionary aspects of extracutaneous melanocytes and found that pigment cells are associated mainly with nerves and blood vessels in amphibians and fish. This new knowledge of the nerve-dependent origin of extracutaneous pigment cells might be directly relevant to the formation of extracutaneous melanoma in humans.


Brain/physiology , Ear, Inner/physiology , Heart/physiology , Meninges/physiology , Nervous System/physiopathology , Schwann Cells/physiology , Amphibians/metabolism , Amphibians/physiology , Animals , Brain/metabolism , Cell Lineage/physiology , Ear, Inner/metabolism , Embryonic Development/physiology , Female , Fishes/metabolism , Fishes/physiology , Melanocytes/metabolism , Melanocytes/physiology , Meninges/metabolism , Mice , Nervous System/metabolism , Pregnancy , Receptor, Endothelin B/metabolism , Schwann Cells/metabolism
11.
Gigascience ; 10(3)2021 03 02.
Article En | MEDLINE | ID: mdl-33677535

BACKGROUND: X-ray microtomography (µCT) has become an invaluable tool for non-destructive analysis of biological samples in the field of developmental biology. Mouse embryos are a typical model for investigation of human developmental diseases. By obtaining 3D high-resolution scans of the mouse embryo heads, we gain valuable morphological information about the structures prominent in the development of future face, brain, and sensory organs. The development of facial skeleton tracked in these µCT data provides a valuable background for further studies of congenital craniofacial diseases and normal development. FINDINGS: In this work, reusable tomographic data from 7 full 3D scans of mouse embryo heads are presented and made publicly available. The ages of these embryos range from E12.5 to E18.5. The samples were stained by phosphotungstic acid prior to scanning, which greatly enhanced the contrast of various tissues in the reconstructed images and enabled precise segmentation. The images were obtained on a laboratory-based µCT system. Furthermore, we provide manually segmented masks of mesenchymal condensations (for E12.5 and E13.5) and cartilage present in the nasal capsule of the scanned embryos. CONCLUSION: We present a comprehensive dataset of X-ray 3D computed tomography images of the developing mouse head with high-quality manual segmentation masks of cartilaginous nasal capsules. The provided µCT images can be used for studying any other major structure within the developing mouse heads. The high quality of the manually segmented models of nasal capsules may be instrumental to understanding the complex process of the development of the face in a mouse model.


Imaging, Three-Dimensional , Skull , Animals , Mice , Skull/diagnostic imaging , X-Ray Microtomography
12.
Dev Dyn ; 250(9): 1300-1317, 2021 09.
Article En | MEDLINE | ID: mdl-33511716

BACKGROUND: In vertebrates, the skull evolves from a complex network of dermal bones and cartilage-the latter forming the pharyngeal apparatus and the chondrocranium. Squamates are particularly important in this regard as they maintain at least part of the chondrocranium throughout their whole ontogeny until adulthood. Anguid lizards represent a unique group of squamates, which contains limbed and limbless forms and show conspicuous variation of the adult skull. RESULTS: Based on several emboadryonic stages of the limbless lizards Pseudopus apodus and Anguis fragilis, and by comparing with other squamates, we identified and interpreted major differences in chondrocranial anatomy. Among others, the most important differences are in the orbitotemporal region. P. apodus shows a strikingly similar development of this region to other squamates. Unexpectedly, however, A. fragilis differs considerably in the composition of the orbitotemporal region. In addition, A. fragilis retains a paedomorphic state of the nasal region. CONCLUSIONS: Taxonomic comparisons indicate that even closely related species with reduced limbs show significant differences in chondrocranial anatomy. The Pearson correlation coefficient suggests strong correlation between chondrocranial reduction and limb reduction. We pose the hypothesis that limb reduction could be associated with the reduction in chondrocrania by means of genetic mechanisms.


Biological Evolution , Lizards , Animals , Cartilage , Extremities , Skull/anatomy & histology
13.
Proc Natl Acad Sci U S A ; 117(30): 17854-17863, 2020 07 28.
Article En | MEDLINE | ID: mdl-32647059

Pacemaker neurons exert control over neuronal circuit function by their intrinsic ability to generate rhythmic bursts of action potential. Recent work has identified rhythmic gut contractions in human, mice, and hydra to be dependent on both neurons and the resident microbiota. However, little is known about the evolutionary origin of these neurons and their interaction with microbes. In this study, we identified and functionally characterized prototypical ANO/SCN/TRPM ion channel-expressing pacemaker cells in the basal metazoan Hydra by using a combination of single-cell transcriptomics, immunochemistry, and functional experiments. Unexpectedly, these prototypical pacemaker neurons express a rich set of immune-related genes mediating their interaction with the microbial environment. Furthermore, functional experiments gave a strong support to a model of the evolutionary emergence of pacemaker cells as neurons using components of innate immunity to interact with the microbial environment and ion channels to generate rhythmic contractions.


Biological Clocks , Hydra/physiology , Microbiota , Neurons/physiology , Action Potentials , Animals , Biological Evolution , Cluster Analysis , Computational Biology/methods , Gene Expression Profiling , Gene Expression Regulation , Genome-Wide Association Study , Humans , Mice
15.
Front Cell Dev Biol ; 8: 620735, 2020.
Article En | MEDLINE | ID: mdl-33392208

The head represents the most complex part of the body and a distinctive feature of the vertebrate body plan. This intricate structure is assembled during embryonic development in the four-dimensional process of morphogenesis. The head integrates components of the central and peripheral nervous system, sensory organs, muscles, joints, glands, and other specialized tissues in the framework of a complexly shaped skull. The anterior part of the head is referred to as the face, and a broad spectrum of facial shapes across vertebrate species enables different feeding strategies, communication styles, and diverse specialized functions. The face formation starts early during embryonic development and is an enormously complex, multi-step process regulated on a genomic, molecular, and cellular level. In this review, we will discuss recent discoveries that revealed new aspects of facial morphogenesis from the time of the neural crest cell emergence till the formation of the chondrocranium, the primary design of the individual facial shape. We will focus on molecular mechanisms of cell fate specification, the role of individual and collective cell migration, the importance of dynamic and continuous cellular interactions, responses of cells and tissues to generated physical forces, and their morphogenetic outcomes. In the end, we will examine the spatiotemporal activity of signaling centers tightly regulating the release of signals inducing the formation of craniofacial skeletal elements. The existence of these centers and their regulation by enhancers represent one of the core morphogenetic mechanisms and might lay the foundations for intra- and inter-species facial variability.

16.
J Vis Exp ; (152)2019 10 23.
Article En | MEDLINE | ID: mdl-31710038

Labeling an individual cell in the body to monitor which cell types it can give rise to and track its migration through the organism or determine its longevity can be a powerful way to reveal mechanisms of tissue development and maintenance. One of the most important tools currently available to monitor cells in vivo is the Confetti mouse model. The Confetti model can be used to genetically label individual cells in living mice with various fluorescent proteins in a cell type-specific manner and monitor their fate, as well as the fate of their progeny over time, in a process called clonal genetic tracing or clonal lineage tracing. This model was generated almost a decade ago and has contributed to an improved understanding of many biological processes, particularly related to stem cell biology, development, and renewal of adult tissues. However, preserving the fluorescent signal until image collection and simultaneous capturing of various fluorescent signals is technically challenging, particularly for mineralized tissue. This publication describes a step-by-step protocol for using the Confetti model to analyze growth plate cartilage that can be applied to any mineralized or nonmineralized tissue.


Calcification, Physiologic/physiology , Cell Lineage , Growth Plate/cytology , Growth Plate/growth & development , Animals , Calcification, Physiologic/genetics , Cells, Cultured , Genes, Reporter , Indicators and Reagents , Mice , Models, Animal , Stem Cells/metabolism
17.
Sci Rep ; 9(1): 14896, 2019 10 17.
Article En | MEDLINE | ID: mdl-31624273

3D imaging approaches based on X-ray microcomputed tomography (microCT) have become increasingly accessible with advancements in methods, instruments and expertise. The synergy of material and life sciences has impacted biomedical research by proposing new tools for investigation. However, data sharing remains challenging as microCT files are usually in the range of gigabytes and require specific and expensive software for rendering and interpretation. Here, we provide an advanced method for visualisation and interpretation of microCT data with small file formats, readable on all operating systems, using freely available Portable Document Format (PDF) software. Our method is based on the conversion of volumetric data into interactive 3D PDF, allowing rotation, movement, magnification and setting modifications of objects, thus providing an intuitive approach to analyse structures in a 3D context. We describe the complete pipeline from data acquisition, data processing and compression, to 3D PDF formatting on an example of craniofacial anatomical morphology in the mouse embryo. Our procedure is widely applicable in biological research and can be used as a framework to analyse volumetric data from any research field relying on 3D rendering and CT-biomedical imaging.


Imaging, Three-Dimensional/statistics & numerical data , Software , X-Ray Microtomography/statistics & numerical data , Animals , Data Compression/statistics & numerical data , Electronic Data Processing , Facial Bones/anatomy & histology , Facial Bones/embryology , Information Dissemination/methods , Mice , Models, Anatomic , Radiographic Image Interpretation, Computer-Assisted , Skull/anatomy & histology , Skull/embryology
18.
Stem Cell Res ; 40: 101563, 2019 10.
Article En | MEDLINE | ID: mdl-31494448

Development of neural tube has been extensively modeled in vitro using human pluripotent stem cells (hPSCs) that are able to form radially organized cellular structures called neural rosettes. While a great amount of research has been done using neural rosettes, studies have only inadequately addressed how rosettes are formed and what the molecular mechanisms and pathways involved in their formation are. Here we address this question by detailed analysis of the expression of pluripotency and differentiation-associated proteins during the early onset of differentiation of hPSCs towards neural rosettes. Additionally, we show that the BMP signaling is likely contributing to the formation of the complex cluster of neural rosettes and its inhibition leads to the altered expression of PAX6, SOX2 and SOX1 proteins and the rosette morphology. Finally, we provide evidence that the mechanism of neural rosettes formation in vitro is reminiscent of the process of secondary neurulation rather than that of primary neurulation in vivo. Since secondary neurulation is a largely unexplored process, its understanding will ultimately assist the development of methods to prevent caudal neural tube defects in humans.


Cell Differentiation , Neural Stem Cells/cytology , Neural Tube/embryology , Neurulation , Pluripotent Stem Cells/cytology , COUP Transcription Factor II/genetics , COUP Transcription Factor II/metabolism , Cells, Cultured , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Neural Stem Cells/metabolism , Neural Tube/cytology , Neural Tube/metabolism , PAX6 Transcription Factor/genetics , PAX6 Transcription Factor/metabolism , POU Domain Factors/genetics , POU Domain Factors/metabolism , Pluripotent Stem Cells/metabolism
19.
Proc Natl Acad Sci U S A ; 116(30): 15068-15073, 2019 07 23.
Article En | MEDLINE | ID: mdl-31285319

Immature multipotent embryonic peripheral glial cells, the Schwann cell precursors (SCPs), differentiate into melanocytes, parasympathetic neurons, chromaffin cells, and dental mesenchymal populations. Here, genetic lineage tracing revealed that, during murine embryonic development, some SCPs detach from nerve fibers to become mesenchymal cells, which differentiate further into chondrocytes and mature osteocytes. This occurred only during embryonic development, producing numerous craniofacial and trunk skeletal elements, without contributing to development of the appendicular skeleton. Formation of chondrocytes from SCPs also occurred in zebrafish, indicating evolutionary conservation. Our findings reveal multipotency of SCPs, providing a developmental link between the nervous system and skeleton.


Bone and Bones/cytology , Cell Lineage/genetics , Chondrocytes/cytology , Mesenchymal Stem Cells/cytology , Nerve Tissue/cytology , Schwann Cells/cytology , Animals , Biomarkers/metabolism , Bone and Bones/embryology , Bone and Bones/metabolism , Cell Differentiation , Chondrocytes/metabolism , Chromaffin Cells/cytology , Chromaffin Cells/metabolism , Embryo, Mammalian , Embryo, Nonmammalian , Embryonic Development , Gene Expression , Melanocytes/cytology , Melanocytes/metabolism , Mesenchymal Stem Cells/metabolism , Mice , Multipotent Stem Cells/cytology , Multipotent Stem Cells/metabolism , Myelin Proteolipid Protein/genetics , Myelin Proteolipid Protein/metabolism , Nerve Fibers/metabolism , Nerve Tissue/embryology , Nerve Tissue/metabolism , Neural Crest/cytology , Neural Crest/growth & development , Neural Crest/metabolism , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Neuroglia/cytology , Neuroglia/metabolism , Neurons/cytology , Neurons/metabolism , Osteocytes/cytology , Osteocytes/metabolism , SOXE Transcription Factors/genetics , SOXE Transcription Factors/metabolism , Schwann Cells/metabolism , Zebrafish/embryology , Zebrafish/genetics , Zebrafish/metabolism
20.
Science ; 364(6444)2019 Jun 07.
Article En | MEDLINE | ID: mdl-31171666

Neural crest cells are embryonic progenitors that generate numerous cell types in vertebrates. With single-cell analysis, we show that mouse trunk neural crest cells become biased toward neuronal lineages when they delaminate from the neural tube, whereas cranial neural crest cells acquire ectomesenchyme potential dependent on activation of the transcription factor Twist1. The choices that neural crest cells make to become sensory, glial, autonomic, or mesenchymal cells can be formalized as a series of sequential binary decisions. Each branch of the decision tree involves initial coactivation of bipotential properties followed by gradual shifts toward commitment. Competing fate programs are coactivated before cells acquire fate-specific phenotypic traits. Determination of a specific fate is achieved by increased synchronization of relevant programs and concurrent repression of competing fate programs.


Gene Expression Regulation, Developmental , Mesenchymal Stem Cells/cytology , Neural Crest/cytology , Neural Crest/embryology , Neural Stem Cells/cytology , Neurogenesis/genetics , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Lineage , Mesenchymal Stem Cells/metabolism , Mice , Mice, Mutant Strains , Nerve Tissue Proteins/metabolism , Neural Crest/metabolism , Neural Stem Cells/metabolism , Neural Tube/cytology , Neural Tube/embryology , Neuroglia/cytology , Neurons/cytology , Nuclear Proteins/metabolism , Single-Cell Analysis , Twist-Related Protein 1/metabolism
...