Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.014
Filter
1.
Nat Commun ; 15(1): 4632, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38951500

ABSTRACT

ANKRD11 (Ankyrin Repeat Domain 11) is a chromatin regulator and a causative gene for KBG syndrome, a rare developmental disorder characterized by multiple organ abnormalities, including cardiac defects. However, the role of ANKRD11 in heart development is unknown. The neural crest plays a leading role in embryonic heart development, and its dysfunction is implicated in congenital heart defects. We demonstrate that conditional knockout of Ankrd11 in the murine embryonic neural crest results in persistent truncus arteriosus, ventricular dilation, and impaired ventricular contractility. We further show these defects occur due to aberrant cardiac neural crest cell organization leading to outflow tract septation failure. Lastly, knockout of Ankrd11 in the neural crest leads to impaired expression of various transcription factors, chromatin remodelers and signaling pathways, including mTOR, BMP and TGF-ß in the cardiac neural crest cells. In this work, we identify Ankrd11 as a regulator of neural crest-mediated heart development and function.


Subject(s)
Heart Defects, Congenital , Heart , Mice, Knockout , Neural Crest , Repressor Proteins , Animals , Female , Mice , Chromatin/metabolism , Gene Expression Regulation, Developmental , Heart/embryology , Heart Defects, Congenital/genetics , Heart Defects, Congenital/metabolism , Heart Defects, Congenital/pathology , Myocardium/metabolism , Neural Crest/metabolism , Neural Crest/embryology , Repressor Proteins/metabolism , Repressor Proteins/genetics , Signal Transduction
2.
Int J Dev Biol ; 68(2): 65-78, 2024 Jul 09.
Article in English | MEDLINE | ID: mdl-39016374

ABSTRACT

During embryonic development, the vertebrate embryonic epiblast is divided into two parts including neural and superficial ectoderm. The neural plate border (NPB) is a narrow transitional area which locates between these parts and contains multipotent progenitor cells. Despite its small size, the cellular heterogeneity in this region produces specific differentiated cells. Signaling pathways, transcription factors, and the expression/repression of certain genes are directly involved in these differentiation processes. Different factors such as the Wnt signaling cascade, fibroblast growth factor (FGF), bone morphogenetic protein (BMP) signaling, and Notch, which are involved in various stages of the growth, proliferation, and differentiation of embryonic cells, are also involved in the determination and differentiation of neural plate border stem cells. Therefore, it is essential to consider the interactions and temporospatial coordination related to cells, tissues, and adjacent structures. This review examines our present knowledge of the formation of the neural plate border and emphasizes the requirement for interaction between different signaling pathways, including the BMP and Wnt cascades, the expression of its special target genes and their regulations, and the precise tissue crosstalk which defines the neural crest fate in the ectoderm at the early human embryonic stages.


Subject(s)
Bone Morphogenetic Proteins , Cell Differentiation , Gene Expression Regulation, Developmental , Neural Crest , Neural Plate , Signal Transduction , Neural Plate/metabolism , Neural Plate/embryology , Humans , Animals , Bone Morphogenetic Proteins/metabolism , Neural Crest/metabolism , Neural Crest/embryology , Ectoderm/metabolism , Ectoderm/embryology , Ectoderm/cytology , Wnt Signaling Pathway/physiology , Fibroblast Growth Factors/metabolism , Fibroblast Growth Factors/genetics , Germ Layers/metabolism , Germ Layers/cytology , Wnt Proteins/metabolism , Wnt Proteins/genetics
3.
Biol Open ; 13(6)2024 Jun 15.
Article in English | MEDLINE | ID: mdl-38874999

ABSTRACT

The neural crest (NC) is an embryonic multipotent and transitory population of cells that appears during late gastrulation/early neurulation in the developing embryos of vertebrate organisms. Often called "the fourth germ layer", the NC is characterised by incredible mobility, which allows the NC cells to migrate throughout the whole embryo, giving rise to an astonishing number of different derivatives in the adult organism, such as craniofacial skeleton, adrenal gland, enteric nervous system and melanocytes. Because of these properties, neurocristopathies (NCPs), which is the term used to classify genetic diseases associated with NC developmental defects, are often syndromic and, taken all together, are the most common type of genetic disease. The NEUcrest consortium is an EU funded innovative training network (ITN) that aims to study the NC and NCPs. In March 2024, the early stage researchers (ESRs) in the NEUcrest consortium organised an in-person conference for well-established and early career researchers to discuss new advances in the NC and NCPs field, starting from the induction of the NC, and then moving on to migration and differentiation processes they undergo. The conference focused heavily on NCPs associated with each of these steps. The conference also included events, such as a round table to discuss the future of the NC research, plus a talk by a person living with an NCP. This 3-day conference aimed to bring together the past, present and future of this field to try and unravel the mysteries of this unique cell population.


Subject(s)
Neural Crest , Animals , Humans , Cell Differentiation , Cell Movement , Neural Crest/cytology , Neural Crest/embryology
4.
Adv Exp Med Biol ; 1441: 125-143, 2024.
Article in English | MEDLINE | ID: mdl-38884708

ABSTRACT

This chapter discusses the role of cardiac neural crest cells in the formation of the septum that divides the cardiac arterial pole into separate systemic and pulmonary arteries. Further, cardiac neural crest cells directly support the normal development and patterning of derivatives of the caudal pharyngeal arches, including the great arteries, thymus, thyroid, and parathyroids. Recently, cardiac neural crest cells have also been shown to indirectly influence the development of the secondary heart field, another derivative of the caudal pharynx, by modulating signaling in the pharynx. The contribution and function of the cardiac neural crest cells has been learned in avian models; most of the genes associated with cardiac neural crest function have been identified using mouse models. Together these studies show that the neural crest cells may not only critical for normal cardiovascular development but also may be involved secondarily because they represent a major component in the complex tissue interactions in the caudal pharynx and outflow tract. Cardiac neural crest cells span from the caudal pharynx into the outflow tract, and therefore may be susceptible to any perturbation in or by other cells in these regions. Thus, understanding congenital cardiac outflow malformations in human sequences of malformations resulting from genetic and/or environmental insults necessarily requires better understanding the role of cardiac neural crest cells in cardiac development.


Subject(s)
Neural Crest , Neural Crest/embryology , Neural Crest/cytology , Neural Crest/metabolism , Animals , Humans , Heart/embryology , Mice
5.
Adv Exp Med Biol ; 1441: 645-659, 2024.
Article in English | MEDLINE | ID: mdl-38884739

ABSTRACT

Tetralogy of Fallot and double-outlet right ventricle are outflow tract (OFT) alignment defects situated on a continuous disease spectrum. A myriad of upstream causes can impact on ventriculoarterial alignment that can be summarized as defects in either i) OFT elongation during looping morphogenesis or ii) OFT remodeling during cardiac septation. Embryological processes underlying these two developmental steps include deployment of second heart field cardiac progenitor cells, establishment and transmission of embryonic left/right information driving OFT rotation and OFT cushion and valve morphogenesis. The formation and remodeling of pulmonary trunk infundibular myocardium is a critical component of both steps. Defects in myocardial, endocardial, or neural crest cell lineages can result in alignment defects, reflecting the complex intercellular signaling events that coordinate arterial pole development. Importantly, however, OFT alignment is mechanistically distinct from neural crest-driven OFT septation, although neural crest cells impact indirectly on alignment through their role in modulating signaling during SHF development. As yet poorly understood nongenetic causes of alignment defects that impact the above processes include hemodynamic changes, maternal exposure to environmental teratogens, and stochastic events. The heterogeneity of causes converging on alignment defects characterizes the OFT as a hotspot of congenital heart defects.


Subject(s)
Disease Models, Animal , Double Outlet Right Ventricle , Signal Transduction , Tetralogy of Fallot , Tetralogy of Fallot/genetics , Tetralogy of Fallot/pathology , Tetralogy of Fallot/physiopathology , Tetralogy of Fallot/embryology , Animals , Double Outlet Right Ventricle/genetics , Double Outlet Right Ventricle/pathology , Double Outlet Right Ventricle/physiopathology , Humans , Neural Crest/metabolism , Neural Crest/pathology , Neural Crest/embryology , Morphogenesis/genetics
6.
Mol Cells ; 47(6): 100076, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38825188

ABSTRACT

The actin-based cytoskeleton is considered a fundamental driving force for cell differentiation and development. Destrin (Dstn), a member of the actin-depolymerizing factor family, regulates actin dynamics by treadmilling actin filaments and increasing globular actin pools. However, the specific developmental roles of dstn have yet to be fully elucidated. Here, we investigated the physiological functions of dstn during early embryonic development using Xenopus laevis as an experimental model organism. dstn is expressed in anterior neural tissue and neural plate during Xenopus embryogenesis. Depleting dstn promoted morphants with short body axes and small heads. Moreover, dstn inhibition extended the neural plate region, impairing cell migration and distribution during neurulation. In addition to the neural plate, dstn knockdown perturbed neural crest cell migration. Our data suggest new insights for understanding the roles of actin dynamics in embryonic neural development, simultaneously presenting a new challenge for studying the complex networks governing cell migration involving actin dynamics.


Subject(s)
Cell Movement , Destrin , Embryonic Development , Xenopus laevis , Animals , Xenopus laevis/embryology , Xenopus laevis/metabolism , Destrin/metabolism , Destrin/genetics , Xenopus Proteins/metabolism , Xenopus Proteins/genetics , Neural Crest/metabolism , Neural Crest/embryology , Neural Crest/cytology , Neurogenesis , Neural Plate/metabolism , Neural Plate/embryology , Actins/metabolism , Gene Expression Regulation, Developmental
7.
Curr Top Dev Biol ; 159: 132-167, 2024.
Article in English | MEDLINE | ID: mdl-38729675

ABSTRACT

The primary senses-touch, taste, sight, smell, and hearing-connect animals with their environments and with one another. Aside from the eyes, the primary sense organs of vertebrates and the peripheral sensory pathways that relay their inputs arise from two transient stem cell populations: the neural crest and the cranial placodes. In this chapter we consider the senses from historical and cultural perspectives, and discuss the senses as biological faculties. We begin with the embryonic origin of the neural crest and cranial placodes from within the neural plate border of the ectodermal germ layer. Then, we describe the major chemical (i.e. olfactory and gustatory) and mechanical (i.e. vestibulo-auditory and somatosensory) senses, with an emphasis on the developmental interactions between neural crest and cranial placodes that shape their structures and functions.


Subject(s)
Neural Crest , Animals , Neural Crest/cytology , Neural Crest/embryology , Neural Crest/physiology , Humans , Sensation/physiology , Sense Organs/embryology , Sense Organs/physiology , Sense Organs/cytology , Vertebrates/embryology , Vertebrates/physiology
8.
Zhonghua Yi Xue Yi Chuan Xue Za Zhi ; 41(6): 758-760, 2024 Jun 10.
Article in Chinese | MEDLINE | ID: mdl-38818565

ABSTRACT

Char syndrome is a rare autosomal dominant genetic disorder characterized by patent ductus arteriosus, facial dysmorphism, and dysplasia of fingers/toes. It may also be associated with multiple papillae, dental dysplasia, and sleep disorders. TFAP2B has proven to be a pathogenic gene for neural crest derivation and development, and several variants of this gene have been identified. Bone morphogenetic protein signaling plays an important role in embryonic development by participating in limb growth and patterning, and regulation of neural crest cell development. TFAP2B is an upstream regulatory gene for bone morphogenetic proteins 2 and 4. Variants of the TFAP2B gene may lead to abnormal proliferation of neural crest cells by affecting the expression of bone morphogenetic proteins, resulting in multiple organ dysplasia syndrome. In addition, TFAP2B variants may only lead to patent ductus arteriosus instead of typical Char syndrome.


Subject(s)
Ductus Arteriosus, Patent , Humans , Ductus Arteriosus, Patent/genetics , Transcription Factor AP-2/genetics , Abnormalities, Multiple/genetics , Bone Morphogenetic Proteins/genetics , Bone Morphogenetic Proteins/metabolism , Neural Crest/metabolism , Neural Crest/embryology , Face/abnormalities , Fingers/abnormalities
9.
Dev Biol ; 511: 63-75, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38621649

ABSTRACT

Loss of function variations in the dual specificity tyrosine-phosphorylation-regulated kinase 1 A (DYRK1A) gene are associated with craniofacial malformations in humans. Here we characterized the effects of deficient DYRK1A in craniofacial development using a developmental model, Xenopus laevis. Dyrk1a mRNA and protein were expressed throughout the developing head and both were enriched in the branchial arches which contribute to the face and jaw. Consistently, reduced Dyrk1a function, using dyrk1a morpholinos and pharmacological inhibitors, resulted in orofacial malformations including hypotelorism, altered mouth shape, slanted eyes, and narrower face accompanied by smaller jaw cartilage and muscle. Inhibition of Dyrk1a function resulted in misexpression of key craniofacial regulators including transcription factors and members of the retinoic acid signaling pathway. Two such regulators, sox9 and pax3 are required for neural crest development and their decreased expression corresponds with smaller neural crest domains within the branchial arches. Finally, we determined that the smaller size of the faces, jaw elements and neural crest domains in embryos deficient in Dyrk1a could be explained by increased cell death and decreased proliferation. This study is the first to provide insight into why craniofacial birth defects might arise in humans with variants of DYRK1A.


Subject(s)
Dyrk Kinases , Xenopus Proteins , Xenopus laevis , Animals , Branchial Region/embryology , Branchial Region/metabolism , Craniofacial Abnormalities/genetics , Craniofacial Abnormalities/embryology , Craniofacial Abnormalities/metabolism , Embryo, Nonmammalian/metabolism , Embryo, Nonmammalian/embryology , Gene Expression Regulation, Developmental , Neural Crest/embryology , Neural Crest/metabolism , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/genetics , Protein-Tyrosine Kinases/metabolism , Protein-Tyrosine Kinases/genetics , Signal Transduction , Xenopus laevis/embryology , Xenopus laevis/metabolism , Xenopus Proteins/metabolism , Xenopus Proteins/genetics
10.
Nat Ecol Evol ; 8(6): 1154-1164, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38565680

ABSTRACT

Neural-crest cells and neuromesodermal progenitors (NMPs) are multipotent cells that are important for development of vertebrate embryos. In embryos of ascidians, which are the closest invertebrate relatives of vertebrates, several cells located at the border between the neural plate and the epidermal region have neural-crest-like properties; hence, the last common ancestor of ascidians and vertebrates may have had ancestral cells similar to neural-crest cells. However, these ascidian neural-crest-like cells do not produce cells that are commonly of mesodermal origin. Here we showed that a cell population located in the lateral region of the neural plate has properties resembling those of vertebrate neural-crest cells and NMPs. Among them, cells with Tbx6-related expression contribute to muscle near the tip of the tail region and cells with Sox1/2/3 expression give rise to the nerve cord. These observations and cross-species transcriptome comparisons indicate that these cells have properties similar to those of NMPs. Meanwhile, transcription factor genes Dlx.b, Zic-r.b and Snai, which are reminiscent of a gene circuit in vertebrate neural-crest cells, are involved in activation of Tbx6-related.b. Thus, the last common ancestor of ascidians and vertebrates may have had cells with properties of neural-crest cells and NMPs and such ancestral cells may have produced cells commonly of ectodermal and mesodermal origins.


Subject(s)
Neural Crest , Vertebrates , Animals , Vertebrates/embryology , Neural Crest/cytology , Neural Crest/embryology , Urochordata/embryology , Urochordata/cytology , Embryo, Nonmammalian/cytology , Ciona intestinalis/embryology , Ciona intestinalis/genetics , Ciona intestinalis/cytology
11.
Elife ; 122024 Apr 18.
Article in English | MEDLINE | ID: mdl-38634469

ABSTRACT

We previously showed that SerpinE2 and the serine protease HtrA1 modulate fibroblast growth factor (FGF) signaling in germ layer specification and head-to-tail development of Xenopus embryos. Here, we present an extracellular proteolytic mechanism involving this serpin-protease system in the developing neural crest (NC). Knockdown of SerpinE2 by injected antisense morpholino oligonucleotides did not affect the specification of NC progenitors but instead inhibited the migration of NC cells, causing defects in dorsal fin, melanocyte, and craniofacial cartilage formation. Similarly, overexpression of the HtrA1 protease impaired NC cell migration and the formation of NC-derived structures. The phenotype of SerpinE2 knockdown was overcome by concomitant downregulation of HtrA1, indicating that SerpinE2 stimulates NC migration by inhibiting endogenous HtrA1 activity. SerpinE2 binds to HtrA1, and the HtrA1 protease triggers degradation of the cell surface proteoglycan Syndecan-4 (Sdc4). Microinjection of Sdc4 mRNA partially rescued NC migration defects induced by both HtrA1 upregulation and SerpinE2 downregulation. These epistatic experiments suggest a proteolytic pathway by a double inhibition mechanism.SerpinE2 ┤HtrA1 protease ┤Syndecan-4 → NC cell migration.


Subject(s)
High-Temperature Requirement A Serine Peptidase 1 , Neural Crest , Serpin E2 , Animals , Cell Movement/genetics , Fibroblast Growth Factors/metabolism , High-Temperature Requirement A Serine Peptidase 1/metabolism , Neural Crest/embryology , Neural Crest/metabolism , Serpin E2/metabolism , Signal Transduction , Xenopus laevis/metabolism , Xenopus Proteins/genetics , Xenopus Proteins/metabolism
12.
Dev Biol ; 511: 26-38, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38580174

ABSTRACT

In a developing embryo, formation of tissues and organs is remarkably precise in both time and space. Through cell-cell interactions, neighboring progenitors coordinate their activities, sequentially generating distinct types of cells. At present, we only have limited knowledge, rather than a systematic understanding, of the underlying logic and mechanisms responsible for cell fate transitions. The formation of the dorsal aspect of the spinal cord is an outstanding model to tackle these dynamics, as it first generates the peripheral nervous system and is later responsible for transmitting sensory information from the periphery to the brain and for coordinating local reflexes. This is reflected first by the ontogeny of neural crest cells, progenitors of the peripheral nervous system, followed by formation of the definitive roof plate of the central nervous system and specification of adjacent interneurons, then a transformation of roof plate into dorsal radial glia and ependyma lining the forming central canal. How do these peripheral and central neural branches segregate from common progenitors? How are dorsal radial glia established concomitant with transformation of the neural tube lumen into a central canal? How do the dorsal radial glia influence neighboring cells? This is only a partial list of questions whose clarification requires the implementation of experimental paradigms in which precise control of timing is crucial. Here, we outline some available answers and still open issues, while highlighting the contributions of avian models and their potential to address mechanisms of neural patterning and function.


Subject(s)
Neural Tube , Spinal Cord , Animals , Spinal Cord/embryology , Neural Tube/embryology , Neural Crest/embryology , Neural Crest/cytology , Neural Crest/physiology , Cell Differentiation/physiology , Neuroglia/physiology , Neuroepithelial Cells/cytology , Neuroepithelial Cells/physiology , Humans
13.
Gastroenterology ; 166(6): 1085-1099, 2024 06.
Article in English | MEDLINE | ID: mdl-38452824

ABSTRACT

BACKGROUND & AIMS: The enteric nervous system (ENS), the gut's intrinsic nervous system critical for gastrointestinal function and gut-brain communication, is believed to mainly originate from vagal neural crest cells (vNCCs) and partially from sacral NCCs (sNCCs). Resolving the exact origins of the ENS is critical for understanding congenital ENS diseases but has been confounded by the inability to distinguish between both NCC populations in situ. Here, we aimed to resolve the exact origins of the mammalian ENS. METHODS: We genetically engineered mouse embryos facilitating comparative lineage-tracing of either all (pan-) NCCs including vNCCs or caudal trunk and sNCCs (s/tNCCs) excluding vNCCs. This was combined with dual-lineage tracing and 3-dimensional reconstruction of pelvic plexus and hindgut to precisely pinpoint sNCC and vNCC contributions. We further used coculture assays to determine the specificity of cell migration from different neural tissues into the hindgut. RESULTS: Both pan-NCCs and s/tNCCs contributed to established NCC derivatives but only pan-NCCs contributed to the ENS. Dual-lineage tracing combined with 3-dimensional reconstruction revealed that s/tNCCs settle in complex patterns in pelvic plexus and hindgut-surrounding tissues, explaining previous confusion regarding their contributions. Coculture experiments revealed unspecific cell migration from autonomic, sensory, and neural tube explants into the hindgut. Lineage tracing of ENS precursors lastly provided complimentary evidence for an exclusive vNCC origin of the murine ENS. CONCLUSIONS: sNCCs do not contribute to the murine ENS, suggesting that the mammalian ENS exclusively originates from vNCCs. These results have immediate implications for comprehending (and devising treatments for) congenital ENS disorders, including Hirschsprung's disease.


Subject(s)
Cell Lineage , Cell Movement , Enteric Nervous System , Neural Crest , Animals , Neural Crest/cytology , Neural Crest/embryology , Enteric Nervous System/embryology , Mice , Coculture Techniques , Mice, Transgenic , Vagus Nerve/embryology , Sacrum/innervation
14.
Nature ; 628(8007): 391-399, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38408487

ABSTRACT

The human nervous system is a highly complex but organized organ. The foundation of its complexity and organization is laid down during regional patterning of the neural tube, the embryonic precursor to the human nervous system. Historically, studies of neural tube patterning have relied on animal models to uncover underlying principles. Recently, models of neurodevelopment based on human pluripotent stem cells, including neural organoids1-5 and bioengineered neural tube development models6-10, have emerged. However, such models fail to recapitulate neural patterning along both rostral-caudal and dorsal-ventral axes in a three-dimensional tubular geometry, a hallmark of neural tube development. Here we report a human pluripotent stem cell-based, microfluidic neural tube-like structure, the development of which recapitulates several crucial aspects of neural patterning in brain and spinal cord regions and along rostral-caudal and dorsal-ventral axes. This structure was utilized for studying neuronal lineage development, which revealed pre-patterning of axial identities of neural crest progenitors and functional roles of neuromesodermal progenitors and the caudal gene CDX2 in spinal cord and trunk neural crest development. We further developed dorsal-ventral patterned microfluidic forebrain-like structures with spatially segregated dorsal and ventral regions and layered apicobasal cellular organizations that mimic development of the human forebrain pallium and subpallium, respectively. Together, these microfluidics-based neurodevelopment models provide three-dimensional lumenal tissue architectures with in vivo-like spatiotemporal cell differentiation and organization, which will facilitate the study of human neurodevelopment and disease.


Subject(s)
Body Patterning , Microfluidics , Neural Tube , Humans , Cell Culture Techniques, Three Dimensional , Cell Differentiation , Neural Crest/cytology , Neural Crest/embryology , Neural Tube/cytology , Neural Tube/embryology , Pluripotent Stem Cells/cytology , Prosencephalon/cytology , Prosencephalon/embryology , Spinal Cord/cytology , Spinal Cord/embryology
15.
Nature ; 612(7941): 732-738, 2022 12.
Article in English | MEDLINE | ID: mdl-36517595

ABSTRACT

Our understanding of human early development is severely hampered by limited access to embryonic tissues. Due to their close evolutionary relationship with humans, nonhuman primates are often used as surrogates to understand human development but currently suffer from a lack of in vivo datasets, especially from gastrulation to early organogenesis during which the major embryonic cell types are dynamically specified. To fill this gap, we collected six Carnegie stage 8-11 cynomolgus monkey (Macaca fascicularis) embryos and performed in-depth transcriptomic analyses of 56,636 single cells. Our analyses show transcriptomic features of major perigastrulation cell types, which help shed light on morphogenetic events including primitive streak development, somitogenesis, gut tube formation, neural tube patterning and neural crest differentiation in primates. In addition, comparative analyses with mouse embryos and human embryoids uncovered conserved and divergent features of perigastrulation development across species-for example, species-specific dependency on Hippo signalling during presomitic mesoderm differentiation-and provide an initial assessment of relevant stem cell models of human early organogenesis. This comprehensive single-cell transcriptome atlas not only fills the knowledge gap in the nonhuman primate research field but also serves as an invaluable resource for understanding human embryogenesis and developmental disorders.


Subject(s)
Gastrulation , Macaca fascicularis , Organogenesis , Single-Cell Analysis , Animals , Humans , Mice , Gastrulation/genetics , Macaca fascicularis/embryology , Macaca fascicularis/genetics , Organogenesis/genetics , Embryoid Bodies , Gene Expression Profiling , Primitive Streak/cytology , Primitive Streak/embryology , Neural Tube/cytology , Neural Tube/embryology , Neural Crest/cytology , Neural Crest/embryology , Hippo Signaling Pathway , Mesoderm/cytology , Mesoderm/embryology , Stem Cells
16.
Nature ; 610(7930): 190-198, 2022 10.
Article in English | MEDLINE | ID: mdl-36131018

ABSTRACT

Although melanoma is notorious for its high degree of heterogeneity and plasticity1,2, the origin and magnitude of cell-state diversity remains poorly understood. Equally, it is unclear whether growth and metastatic dissemination are supported by overlapping or distinct melanoma subpopulations. Here, by combining mouse genetics, single-cell and spatial transcriptomics, lineage tracing and quantitative modelling, we provide evidence of a hierarchical model of tumour growth that mirrors the cellular and molecular logic underlying the cell-fate specification and differentiation of the embryonic neural crest. We show that tumorigenic competence is associated with a spatially localized perivascular niche, a phenotype acquired through an intercellular communication pathway established by endothelial cells. Consistent with a model in which only a fraction of cells are fated to fuel growth, temporal single-cell tracing of a population of melanoma cells with a mesenchymal-like state revealed that these cells do not contribute to primary tumour growth but, instead, constitute a pool of metastatic initiating cells that switch cell identity while disseminating to secondary organs. Our data provide a spatially and temporally resolved map of the diversity and trajectories of melanoma cell states and suggest that the ability to support growth and metastasis are limited to distinct pools of cells. The observation that these phenotypic competencies can be dynamically acquired after exposure to specific niche signals warrant the development of therapeutic strategies that interfere with the cancer cell reprogramming activity of such microenvironmental cues.


Subject(s)
Cell Proliferation , Melanoma , Neoplasm Metastasis , Animals , Cell Communication , Cell Differentiation , Cell Lineage , Cell Tracking , Cellular Reprogramming , Endothelial Cells , Melanoma/genetics , Melanoma/pathology , Mesoderm/pathology , Mice , Neoplasm Metastasis/pathology , Neural Crest/embryology , Phenotype , Single-Cell Analysis , Transcriptome , Tumor Microenvironment
17.
Proc Natl Acad Sci U S A ; 119(31): e2116974119, 2022 08 02.
Article in English | MEDLINE | ID: mdl-35881792

ABSTRACT

Ribosomal RNA (rRNA) transcription by RNA polymerase I (Pol I) is a critical rate-limiting step in ribosome biogenesis, which is essential for cell survival. Despite its global function, disruptions in ribosome biogenesis cause tissue-specific birth defects called ribosomopathies, which frequently affect craniofacial development. Here, we describe a cellular and molecular mechanism underlying the susceptibility of craniofacial development to disruptions in Pol I transcription. We show that Pol I subunits are highly expressed in the neuroepithelium and neural crest cells (NCCs), which generate most of the craniofacial skeleton. High expression of Pol I subunits sustains elevated rRNA transcription in NCC progenitors, which supports their high tissue-specific levels of protein translation, but also makes NCCs particularly sensitive to rRNA synthesis defects. Consistent with this model, NCC-specific deletion of Pol I subunits Polr1a, Polr1c, and associated factor Tcof1 in mice cell-autonomously diminishes rRNA synthesis, which leads to p53 protein accumulation, resulting in NCC apoptosis and craniofacial anomalies. Furthermore, compound mutations in Pol I subunits and associated factors specifically exacerbate the craniofacial anomalies characteristic of the ribosomopathies Treacher Collins syndrome and Acrofacial Dysostosis-Cincinnati type. Mechanistically, we demonstrate that diminished rRNA synthesis causes an imbalance between rRNA and ribosomal proteins. This leads to increased binding of ribosomal proteins Rpl5 and Rpl11 to Mdm2 and concomitantly diminished binding between Mdm2 and p53. Altogether, our results demonstrate a dynamic spatiotemporal requirement for rRNA transcription during mammalian cranial NCC development and corresponding tissue-specific threshold sensitivities to disruptions in rRNA transcription in the pathogenesis of congenital craniofacial disorders.


Subject(s)
Craniofacial Abnormalities , RNA Polymerase I , RNA, Ribosomal , Ribosomal Proteins , Skull , Transcription, Genetic , Animals , Craniofacial Abnormalities/genetics , Mandibulofacial Dysostosis/genetics , Mice , Neural Crest/embryology , Proto-Oncogene Proteins c-mdm2/metabolism , RNA Polymerase I/metabolism , RNA, Ribosomal/genetics , Ribosomal Proteins/metabolism , Skull/embryology , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
18.
Elife ; 112022 01 19.
Article in English | MEDLINE | ID: mdl-35044299

ABSTRACT

Serum response factor (SRF) is an essential transcription factor that influences many cellular processes including cell proliferation, migration, and differentiation. SRF directly regulates and is required for immediate early gene (IEG) and actin cytoskeleton-related gene expression. SRF coordinates these competing transcription programs through discrete sets of cofactors, the ternary complex factors (TCFs) and myocardin-related transcription factors (MRTFs). The relative contribution of these two programs to in vivo SRF activity and mutant phenotypes is not fully understood. To study how SRF utilizes its cofactors during development, we generated a knock-in SrfaI allele in mice harboring point mutations that disrupt SRF-MRTF-DNA complex formation but leave SRF-TCF activity unaffected. Homozygous SrfaI/aI mutants die at E10.5 with notable cardiovascular phenotypes, and neural crest conditional mutants succumb at birth to defects of the cardiac outflow tract but display none of the craniofacial phenotypes associated with complete loss of SRF in that lineage. Our studies further support an important role for MRTF mediating SRF function in cardiac neural crest and suggest new mechanisms by which SRF regulates transcription during development.


Subject(s)
Neural Crest/embryology , Serum Response Factor/genetics , Ternary Complex Factors/genetics , Transcription Factors/genetics , Animals , Mice , Serum Response Factor/metabolism , Ternary Complex Factors/metabolism , Transcription Factors/metabolism
19.
Dev Biol ; 483: 39-57, 2022 03.
Article in English | MEDLINE | ID: mdl-34990731

ABSTRACT

Neural crest (NC) cells are a dynamic population of embryonic stem cells that create various adult tissues in vertebrate species including craniofacial bone and cartilage and the peripheral and enteric nervous systems. NC development is thought to be a conserved and complex process that is controlled by a tightly-regulated gene regulatory network (GRN) of morphogens, transcription factors, and cell adhesion proteins. While multiple studies have characterized the expression of several GRN factors in single species, a comprehensive protein analysis that directly compares expression across development is lacking. To address this lack in information, we used three closely related avian models, Gallus gallus (chicken), Coturnix japonica (Japanese quail), and Pavo cristatus (Indian peafowl), to compare the localization and timing of four GRN transcription factors, PAX7, SNAI2, SOX9, and SOX10, from the onset of neurulation to migration. While the spatial expression of these factors is largely conserved, we find that quail NC cells express SNAI2, SOX9, and SOX10 proteins at the equivalent of earlier developmental stages than chick and peafowl. In addition, quail NC cells migrate farther and more rapidly than the larger organisms. These data suggest that despite a conservation of NC GRN players, differences in the timing of NC development between species remain a significant frontier to be explored with functional studies.


Subject(s)
Avian Proteins/genetics , Avian Proteins/metabolism , Cell Movement/genetics , Chickens/genetics , Coturnix/embryology , Coturnix/genetics , Gene Expression Regulation, Developmental , Neural Crest/metabolism , Neurulation/genetics , Animals , Chick Embryo , Chickens/metabolism , Coturnix/metabolism , Female , Gene Regulatory Networks , Neural Crest/embryology , Neural Tube/embryology , Neural Tube/metabolism , Oviparity/genetics , PAX7 Transcription Factor/genetics , PAX7 Transcription Factor/metabolism , SOX9 Transcription Factor/genetics , SOX9 Transcription Factor/metabolism , Snail Family Transcription Factors/genetics , Snail Family Transcription Factors/metabolism
20.
Elife ; 112022 01 28.
Article in English | MEDLINE | ID: mdl-35088714

ABSTRACT

The epiblast of vertebrate embryos is comprised of neural and non-neural ectoderm, with the border territory at their intersection harboring neural crest and cranial placode progenitors. Here, we a generate single-cell atlas of the developing chick epiblast from late gastrulation through early neurulation stages to define transcriptional changes in the emerging 'neural plate border' as well as other regions of the epiblast. Focusing on the border territory, the results reveal gradual establishment of heterogeneous neural plate border signatures, including novel genes that we validate by fluorescent in situ hybridization. Developmental trajectory analysis infers that segregation of neural plate border lineages only commences at early neurulation, rather than at gastrulation as previously predicted. We find that cells expressing the prospective neural crest marker Pax7 contribute to multiple lineages, and a subset of premigratory neural crest cells shares a transcriptional signature with their border precursors. Together, our results suggest that cells at the neural plate border remain heterogeneous until early neurulation, at which time progenitors become progressively allocated toward defined neural crest and placode lineages. The data also can be mined to reveal changes throughout the developing epiblast.


Subject(s)
Gene Expression Regulation, Developmental , Neural Crest/embryology , Neural Plate/embryology , Neurulation/physiology , Animals , Chick Embryo/cytology , Chickens/physiology , Germ Layers/physiology , In Situ Hybridization, Fluorescence , PAX7 Transcription Factor/analysis
SELECTION OF CITATIONS
SEARCH DETAIL