Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 7.089
Filter
1.
Development ; 151(20)2024 Oct 15.
Article in English | MEDLINE | ID: mdl-39109637

ABSTRACT

Vertebrate calcitonin-producing cells (C-cells) are neuroendocrine cells that secrete the small peptide hormone calcitonin in response to elevated blood calcium levels. Whereas mouse C-cells reside within the thyroid gland and derive from pharyngeal endoderm, avian C-cells are located within ultimobranchial glands and have been reported to derive from the neural crest. We use a comparative cell lineage tracing approach in a range of vertebrate model systems to resolve the ancestral embryonic origin of vertebrate C-cells. We find, contrary to previous studies, that chick C-cells derive from pharyngeal endoderm, with neural crest-derived cells instead contributing to connective tissue intimately associated with C-cells in the ultimobranchial gland. This endodermal origin of C-cells is conserved in a ray-finned bony fish (zebrafish) and a cartilaginous fish (the little skate, Leucoraja erinacea). Furthermore, we discover putative C-cell homologs within the endodermally-derived pharyngeal epithelium of the ascidian Ciona intestinalis and the amphioxus Branchiostoma lanceolatum, two invertebrate chordates that lack neural crest cells. Our findings point to a conserved endodermal origin of C-cells across vertebrates and to a pre-vertebrate origin of this cell type along the chordate stem.


Subject(s)
Calcitonin , Cell Lineage , Ciona intestinalis , Endoderm , Neural Crest , Neuroendocrine Cells , Animals , Endoderm/metabolism , Endoderm/cytology , Calcitonin/metabolism , Neuroendocrine Cells/metabolism , Neuroendocrine Cells/cytology , Ciona intestinalis/metabolism , Ciona intestinalis/embryology , Neural Crest/metabolism , Neural Crest/cytology , Chick Embryo , Mice , Vertebrates/embryology , Vertebrates/metabolism , Zebrafish/embryology , Lancelets/embryology , Lancelets/metabolism , Lancelets/genetics , Ultimobranchial Body/metabolism
2.
Development ; 151(15)2024 Aug 01.
Article in English | MEDLINE | ID: mdl-39092608

ABSTRACT

Melanocytes evolved to produce the melanin that gives colour to our hair, eyes and skin. The melanocyte lineage also gives rise to melanoma, the most lethal form of skin cancer. The melanocyte lineage differentiates from neural crest cells during development, and most melanocytes reside in the skin and hair, where they are replenished by melanocyte stem cells. Because the molecular mechanisms necessary for melanocyte specification, migration, proliferation and differentiation are co-opted during melanoma initiation and progression, studying melanocyte development is directly relevant to human disease. Here, through the lens of advances in cellular omic and genomic technologies, we review the latest findings in melanocyte development and differentiation, and how these developmental pathways become dysregulated in disease.


Subject(s)
Cell Differentiation , Cell Lineage , Melanocytes , Melanoma , Melanocytes/metabolism , Melanocytes/cytology , Humans , Animals , Melanoma/pathology , Melanoma/metabolism , Melanoma/genetics , Neural Crest/metabolism , Cell Proliferation , Skin Neoplasms/pathology , Skin Neoplasms/metabolism , Skin Neoplasms/genetics
3.
Nat Commun ; 15(1): 7065, 2024 Aug 16.
Article in English | MEDLINE | ID: mdl-39152112

ABSTRACT

The sympathetic nervous system controls bodily functions including vascular tone, cardiac rhythm, and the "fight-or-flight response". Sympathetic chain ganglia develop in parallel with preganglionic motor nerves extending from the neural tube, raising the question of whether axon targeting contributes to sympathetic chain formation. Using nerve-selective genetic ablations and lineage tracing in mouse, we reveal that motor nerve-associated Schwann cell precursors (SCPs) contribute sympathetic neurons and satellite glia after the initial seeding of sympathetic ganglia by neural crest. Motor nerve ablation causes mispositioning of SCP-derived sympathoblasts as well as sympathetic chain hypoplasia and fragmentation. Sympathetic neurons in motor-ablated embryos project precociously and abnormally towards dorsal root ganglia, eventually resulting in fusion of sympathetic and sensory ganglia. Cell interaction analysis identifies semaphorins as potential motor nerve-derived signaling molecules regulating sympathoblast positioning and outgrowth. Overall, central innervation functions both as infrastructure and regulatory niche to ensure the integrity of peripheral ganglia morphogenesis.


Subject(s)
Ganglia, Sympathetic , Motor Neurons , Neural Crest , Schwann Cells , Sympathetic Nervous System , Animals , Sympathetic Nervous System/embryology , Mice , Motor Neurons/physiology , Schwann Cells/metabolism , Neural Crest/cytology , Neural Crest/metabolism , Ganglia, Sympathetic/cytology , Ganglia, Spinal , Semaphorins/metabolism , Semaphorins/genetics , Mice, Transgenic , Neuroglia/metabolism , Female
4.
Int J Mol Sci ; 25(15)2024 Jul 23.
Article in English | MEDLINE | ID: mdl-39125577

ABSTRACT

Mesenchymal stromal cells (MSCs) display heterogeneity in origin and functional role in tissue homeostasis. Subsets of MSCs derived from the neural crest express nestin and serve as niches in bone marrow, but the possibility of coaxing MSCs into nestin-expresing cells for enhanced supportive activity is unclear. In this study, as an approach to the chemical coaxing of MSC functions, we screened libraries of clinically approved chemicals to identify compounds capable of inducing nestin expression in MSCs. Out of 2000 clinical compounds, we chose vorinostat as a candidate to coax the MSCs into neural crest-like fates. When treated with vorinostat, MSCs exhibited a significant increase in the expression of genes involved in the pluripotency and epithelial-mesenchymal transition (EMT), as well as nestin and CD146, the markers for pericytes. In addition, these nestin-induced MSCs exhibited enhanced differentiation towards neuronal cells with the upregulation of neurogenic markers, including SRY-box transcription factor 2 (Sox2), SRY-box transcription factor 10 (Sox10) and microtubule associated protein 2 (Map2) in addition to nestin. Moreover, the coaxed MSCs exhibited enhanced supporting activity for hematopoietic progenitors without supporting leukemia cells. These results demonstrate the feasibility of the drug repositioning of MSCs to induce neural crest-like properties through the chemical coaxing of cell fates.


Subject(s)
Cell Differentiation , Drug Repositioning , Mesenchymal Stem Cells , Nestin , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/cytology , Nestin/metabolism , Nestin/genetics , Humans , Cell Differentiation/drug effects , Drug Repositioning/methods , Epithelial-Mesenchymal Transition/drug effects , Cells, Cultured , Neural Crest/cytology , Neural Crest/metabolism , Neural Crest/drug effects
5.
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
6.
Front Biosci (Landmark Ed) ; 29(7): 261, 2024 Jul 22.
Article in English | MEDLINE | ID: mdl-39082344

ABSTRACT

The neural crest (NC), also known as the "fourth germ layer", is an embryonic structure with important contributions to multiple tissue and organ systems. Neural crest cells (NCCs) are subjected to epithelial to mesenchymal transition and migrate throughout the embryo until they reach their destinations, where they differentiate into discrete cell types. Specific gene expression enables this precise NCCs delamination and colonization potency in distinct and diverse locations therein. This review aims to summarize the current experimental evidence from multiple species into the NCCs specifier genes that drive this embryo body axes segmentation. Additionally, it attempts to filter further into the genetic background that produces these individual cell subpopulations. Understanding the multifaceted genetic makeup that shapes NC-related embryonic structures will offer valuable insights to researchers studying organogenesis and disease phenotypes arising from dysmorphogenesis.


Subject(s)
Cell Differentiation , Neural Crest , Organogenesis , Neural Crest/cytology , Animals , Cell Differentiation/genetics , Organogenesis/genetics , Humans , Gene Expression Regulation, Developmental , Epithelial-Mesenchymal Transition/genetics
7.
Int J Mol Sci ; 25(13)2024 Jun 28.
Article in English | MEDLINE | ID: mdl-39000221

ABSTRACT

The traditional description of cardiac development involves progression from a cardiac crescent to a linear heart tube, which in the phase of transformation into a mature heart forms a cardiac loop and is divided with the septa into individual cavities. Cardiac morphogenesis involves numerous types of cells originating outside the initial cardiac crescent, including neural crest cells, cells of the second heart field origin, and epicardial progenitor cells. The development of the fetal heart and circulatory system is subject to regulatation by both genetic and environmental processes. The etiology for cases with congenital heart defects (CHDs) is largely unknown, but several genetic anomalies, some maternal illnesses, and prenatal exposures to specific therapeutic and non-therapeutic drugs are generally accepted as risk factors. New techniques for studying heart development have revealed many aspects of cardiac morphogenesis that are important in the development of CHDs, in particular transposition of the great arteries.


Subject(s)
Heart Defects, Congenital , Heart , Humans , Heart Defects, Congenital/pathology , Heart Defects, Congenital/etiology , Animals , Heart/embryology , Heart/growth & development , Neural Crest , Morphogenesis , Organogenesis
8.
Cell Rep ; 43(7): 114406, 2024 Jul 23.
Article in English | MEDLINE | ID: mdl-38963759

ABSTRACT

Cancer cellular heterogeneity and therapy resistance arise substantially from metabolic and transcriptional adaptations, but how these are interconnected is poorly understood. Here, we show that, in melanoma, the cancer stem cell marker aldehyde dehydrogenase 1A3 (ALDH1A3) forms an enzymatic partnership with acetyl-coenzyme A (CoA) synthetase 2 (ACSS2) in the nucleus to couple high glucose metabolic flux with acetyl-histone H3 modification of neural crest (NC) lineage and glucose metabolism genes. Importantly, we show that acetaldehyde is a metabolite source for acetyl-histone H3 modification in an ALDH1A3-dependent manner, providing a physiologic function for this highly volatile and toxic metabolite. In a zebrafish melanoma residual disease model, an ALDH1-high subpopulation emerges following BRAF inhibitor treatment, and targeting these with an ALDH1 suicide inhibitor, nifuroxazide, delays or prevents BRAF inhibitor drug-resistant relapse. Our work reveals that the ALDH1A3-ACSS2 couple directly coordinates nuclear acetaldehyde-acetyl-CoA metabolism with specific chromatin-based gene regulation and represents a potential therapeutic vulnerability in melanoma.


Subject(s)
Acetaldehyde , Melanoma , Zebrafish , Melanoma/metabolism , Melanoma/genetics , Melanoma/pathology , Melanoma/drug therapy , Acetaldehyde/metabolism , Acetaldehyde/pharmacology , Animals , Humans , Cell Line, Tumor , Aldehyde Oxidoreductases/metabolism , Aldehyde Oxidoreductases/genetics , Histones/metabolism , Coenzyme A Ligases/metabolism , Coenzyme A Ligases/genetics , Transcription, Genetic/drug effects , Neural Crest/metabolism , Neural Crest/drug effects , Gene Expression Regulation, Neoplastic/drug effects
9.
PLoS Biol ; 22(7): e3002074, 2024 Jul.
Article in English | MEDLINE | ID: mdl-39038054

ABSTRACT

While interactions between neural crest and placode cells are critical for the proper formation of the trigeminal ganglion, the mechanisms underlying this process remain largely uncharacterized. Here, by using chick embryos, we show that the microRNA (miR)-203, whose epigenetic repression is required for neural crest migration, is reactivated in coalescing and condensing trigeminal ganglion cells. Overexpression of miR-203 induces ectopic coalescence of neural crest cells and increases ganglion size. By employing cell-specific electroporations for either miR-203 sponging or genomic editing using CRISPR/Cas9, we elucidated that neural crest cells serve as the source, while placode cells serve as the site of action for miR-203 in trigeminal ganglion condensation. Demonstrating intercellular communication, overexpression of miR-203 in the neural crest in vitro or in vivo represses an miR-responsive sensor in placode cells. Moreover, neural crest-secreted extracellular vesicles (EVs), visualized using pHluorin-CD63 vector, become incorporated into the cytoplasm of placode cells. Finally, RT-PCR analysis shows that small EVs isolated from condensing trigeminal ganglia are selectively loaded with miR-203. Together, our findings reveal a critical role in vivo for neural crest-placode communication mediated by sEVs and their selective microRNA cargo for proper trigeminal ganglion formation.


Subject(s)
Cell Communication , Extracellular Vesicles , MicroRNAs , Neural Crest , Trigeminal Ganglion , Neural Crest/metabolism , Neural Crest/embryology , Neural Crest/cytology , Animals , MicroRNAs/metabolism , MicroRNAs/genetics , Trigeminal Ganglion/metabolism , Trigeminal Ganglion/embryology , Trigeminal Ganglion/cytology , Extracellular Vesicles/metabolism , Chick Embryo , Cell Communication/genetics , Cell Movement/genetics , Gene Expression Regulation, Developmental
10.
Life Sci ; 352: 122869, 2024 Sep 01.
Article in English | MEDLINE | ID: mdl-38950644

ABSTRACT

AIMS: To detect the therapeutic efficacy of CelTrac1000-labeled hair follicle epidermal neural crest stem cells (EPI-NCSCs) on repairing facial nerve defects by second near-infrared (NIR-II) fluorescence imaging. MATERIALS AND METHODS: Firstly, CelTrac1000-labeled EPI-NCSCs were microinjected into the acellular nerve allografts (ANAs) to bridge a 10-mm-long gap in the buccal branch of facial nerve in adult rats. Then, Celtrac1000-labeled EPI-NCSCs were detected by NIR-II fluorescence imaging system to visualize the behavior of the transplanted cells in vivo. Additionally, the effect of the transplanted EPI-NCSCs on repairing facial nerve defect was examined. KEY FINDINGS: Through 14 weeks of dynamic observation, the transplanted EPI-NCSCs survived in the ANAs in vivo after surgery. Meanwhile, the region of the NIR-II fluorescence signals was gradually limited to be consistent with the direction of the regenerative nerve segment. Furthermore, the results of functional and morphological analysis showed that the transplanted EPI-NCSCs could promote the recovery of facial paralysis and neural regeneration after injury. SIGNIFICANCE: Our research provides a novel way to track the transplanted cells in preclinical studies of cell therapy for facial paralysis, and demonstrates the therapeutic potential of EPI-NCSCs combined with ANAs in bridging rat facial nerve defects.


Subject(s)
Facial Nerve Injuries , Hair Follicle , Nerve Regeneration , Neural Crest , Neural Stem Cells , Optical Imaging , Animals , Rats , Neural Crest/cytology , Neural Crest/transplantation , Nerve Regeneration/physiology , Facial Nerve Injuries/therapy , Neural Stem Cells/transplantation , Optical Imaging/methods , Rats, Sprague-Dawley , Male , Facial Nerve , Stem Cell Transplantation/methods , Chitosan/chemistry
11.
Development ; 151(13)2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38958074

ABSTRACT

The bromodomain and extra-terminal (BET) family of proteins reads epigenetic histone acetylation marks on the genome and regulates the transcriptional machinery. In their study, Carole LaBonne and colleagues reveal the role of BET protein activity in the maintenance of pluripotency and establishment of the neural crest in Xenopus laevis. To know more about their work, we spoke to the first author Paul Huber and the corresponding author Carole LaBonne, Developmental and Stem Cell Biologist at Northwestern University.


Subject(s)
Xenopus laevis , Animals , History, 21st Century , Humans , History, 20th Century , Neural Crest/metabolism , Developmental Biology/history
12.
Curr Opin Cell Biol ; 89: 102400, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39032482

ABSTRACT

Cells have evolved mechanisms to migrate for diverse biological functions. A process frequently deployed during metazoan cell migration is the epithelial-mesenchymal transition (EMT). During EMT, adherent epithelial cells undergo coordinated cellular transitions to mesenchymalize and reduce their intercellular attachments. This is achieved via tightly regulated changes in gene expression, which modulates cell-cell and cell-matrix adhesion to allow movement. The acquisition of motility and invasive properties following EMT allows some mesenchymal cells to migrate through complex environments to form tissues during embryogenesis; however, these processes may also be leveraged by cancer cells, which often co-opt these endogenous programs to metastasize. Post-transcriptional regulation is now emerging as a major conserved mechanism by which cells modulate EMT and migration, which we discuss here in the context of vertebrate development and cancer.


Subject(s)
Cell Movement , Epithelial-Mesenchymal Transition , Neoplasms , Neural Crest , Neural Crest/metabolism , Neural Crest/cytology , Humans , Animals , Neoplasms/metabolism , Neoplasms/pathology , Neoplasms/genetics , RNA Processing, Post-Transcriptional
13.
Ann N Y Acad Sci ; 1537(1): 113-128, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38970771

ABSTRACT

Goldenhar syndrome, a rare craniofacial malformation, is characterized by developmental anomalies in the first and second pharyngeal arches. Its etiology is considered to be heterogenous, including both genetic and environmental factors that remain largely unknown. To further elucidate the genetic cause in a five-generation Goldenhar syndrome pedigree and exploit the whole-exome sequencing (WES) data of this pedigree, we generated collapsed haplotype pattern markers based on WES and employed rare variant nonparametric linkage analysis. FBLN2 was identified as a candidate gene via analysis of WES data across the significant linkage region. A fbln2 knockout zebrafish line was established by CRISPR/Cas9 to examine the gene's role in craniofacial cartilage development. fbln2 was expressed specifically in the mandible during the zebrafish early development, while fbln2 knockout zebrafish exhibited craniofacial malformations with abnormal chondrocyte morphologies. Functional studies revealed that fbln2 knockout caused abnormal chondrogenic differentiation, apoptosis, and proliferation of cranial neural crest cells (CNCCs), and downregulated the bone morphogenic protein (BMP) signaling pathway in the zebrafish model. This study demonstrates the role of FBLN2 in CNCC development and BMP pathway regulation, and highlights FBLN2 as a candidate gene for Goldenhar syndrome, which may have implications for the selection of potential screening targets and the development of treatments for conditions like microtia-atresia.


Subject(s)
Goldenhar Syndrome , Neural Crest , Pedigree , Zebrafish , Animals , Zebrafish/embryology , Zebrafish/genetics , Neural Crest/metabolism , Goldenhar Syndrome/genetics , Goldenhar Syndrome/metabolism , Goldenhar Syndrome/pathology , Humans , Female , Male , Cell Differentiation/genetics , Exome Sequencing , Chondrogenesis/genetics , Signal Transduction/genetics , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism , Bone Morphogenetic Proteins/metabolism , Bone Morphogenetic Proteins/genetics
14.
Dev Biol ; 515: 79-91, 2024 Nov.
Article in English | MEDLINE | ID: mdl-39019425

ABSTRACT

The trigeminal ganglion, the largest of the vertebrate cranial ganglia, is comprised of sensory neurons that relay sensations of pain, touch, and temperature to the brain. These neurons are derived from two embryonic cell types, the neural crest and ectodermal placodes, whose interactions are critical for proper ganglion formation. While the T-cell leukemia homeobox 3 (Tlx3) gene is known to be expressed in placodally-derived sensory neurons and necessary for their differentiation, little was known about Tlx3 expression and/or function in the neural crest-derived component of the developing trigeminal ganglion. By combining lineage labeling with in situ hybridization in the chick embryo, we show that neural crest-derived cells that contribute to the cranial trigeminal ganglion express Tlx3 at a time point that coincides with the onset of ganglion condensation. Importantly, loss of Tlx3 function in vivo diminishes the overall size and abundance of neurons within the trigeminal ganglion. Conversely, ectopic expression of Tlx3 in migrating cranial neural crest results in their premature neuronal differentiation. Taken together, our results demonstrate a critical role for Tlx3 in neural crest-derived cells during chick trigeminal gangliogenesis.


Subject(s)
Cell Differentiation , Gene Expression Regulation, Developmental , Homeodomain Proteins , Neural Crest , Trigeminal Ganglion , Animals , Trigeminal Ganglion/metabolism , Trigeminal Ganglion/embryology , Trigeminal Ganglion/cytology , Chick Embryo , Neural Crest/metabolism , Neural Crest/embryology , Neural Crest/cytology , Homeodomain Proteins/metabolism , Homeodomain Proteins/genetics , Neurons/metabolism , Neurogenesis/genetics , Cell Movement , Cell Lineage
15.
Dev Biol ; 515: 178-185, 2024 Nov.
Article in English | MEDLINE | ID: mdl-39021074

ABSTRACT

The somatosensory system detects peripheral stimuli that are translated into behaviors necessary for survival. Fishes and amphibians possess two somatosensory systems in the trunk: the primary somatosensory system, formed by the Rohon-Beard neurons, and the secondary somatosensory system, formed by the neural crest cell-derived neurons of the Dorsal Root Ganglia. Rohon-Beard neurons have been characterized as a transient population that mostly disappears during the first days of life and is functionally replaced by the Dorsal Root Ganglia. Here, I follow Rohon-Beard neurons in vivo and show that the entire repertoire remains present in zebrafish from 1-day post-fertilization until the juvenile stage, 15-days post-fertilization. These data indicate that zebrafish retain two complete somatosensory systems until at least a developmental stage when the animals display complex behavioral repertoires.


Subject(s)
Zebrafish , Animals , Zebrafish/embryology , Ganglia, Spinal/embryology , Neurons/physiology , Neural Crest/cytology , Neural Crest/embryology , Neural Crest/physiology
16.
Hum Cell ; 37(5): 1544-1552, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38967841

ABSTRACT

Over the years, the origin of ovarian Leydig cells has been, and still is, a topic subject to deep debate. Seven years ago, we proposed that this origin resided in intraneural elements that came from a possible reservoir of neural crest cells, a reservoir that may be located in the ganglia of the celiac plexus. We believe we have found the evidence necessary to prove this hypothesis.


Subject(s)
Leydig Cells , Ovary , Female , Leydig Cells/cytology , Leydig Cells/physiology , Humans , Ovary/cytology , Animals , Neural Crest/cytology , Ganglia, Sympathetic/cytology
17.
Hua Xi Kou Qiang Yi Xue Za Zhi ; 42(4): 435-443, 2024 Aug 01.
Article in English, Chinese | MEDLINE | ID: mdl-39049630

ABSTRACT

OBJECTIVES: This study aimed to explore the heterogeneity and gene ontology of Wnt1-Cre-marked and Pax2-Cre-marked first branchial arch cranial neural crest cells (CNCs) in mice. METHODS: The embryos of Wnt1-Cre;R26RmTmG and Pax2-Cre;R26RmTmG at embryonic day (E)8.0-E9.25 were collected for histological observation. We performed immunostaining to compare green fluorescent protein (GFP)-positive CNCs in Pax2-Cre;R26RAi9 and Wnt1-Cre;R26RAi9 mice at E15.5. Single-cell RNA sequencing (scRNA-seq) was used to analyze the first branchial arch GFP-positive CNCs from Wnt1-Cre;R26RmTmG and Pax2-cre;R26RmTmGmice at E10.5. Real time fluorescence quantitative polymerase chain reaction (q-PCR) was performed to validate the differential genes. RESULTS: Wnt1-Cre-marked and Pax2-Cre-marked CNCs migrated from the neural plateto first and second branchial arches and to the first branchial arch, respectively, at E8.0. Although Wnt1-Cre-marked and Pax2-Cre-marked CNCs were found mostly in cranial-facial tissues, the former had higher expression in palate and tongue. The results of scRNA-seq showed that Pax2-Cre-marked CNCs specifically contributed to osteoblast differentiation and ossification, while Wnt1-Cre-marked CNCs participated in limb development, cell migration, and ossification. The q-PCR data also confirmed the results of gene ontology analysis. CONCLUSIONS: Pax2-Cre mice are perfect experimental animal models for research on first branchial arch CNCs and derivatives in osteoblast differentiation and ossification.


Subject(s)
Branchial Region , Neural Crest , PAX2 Transcription Factor , Wnt1 Protein , Animals , Neural Crest/metabolism , Mice , Wnt1 Protein/metabolism , PAX2 Transcription Factor/metabolism , Integrases/metabolism , Green Fluorescent Proteins/metabolism
18.
Biofabrication ; 16(4)2024 Aug 14.
Article in English | MEDLINE | ID: mdl-39084624

ABSTRACT

Three-dimensional (3D) tissue models have gained recognition for their improved ability to mimic the native cell microenvironment compared to traditional two-dimensional models. This progress has been driven by advances in tissue-engineering technologies such as 3D bioprinting, a promising method for fabricating biomimetic living tissues. While bioprinting has succeeded in generating various tissues to date, creating neural tissue models remains challenging. In this context, we present an accelerated approach to fabricate 3D sensory neuron (SN) structures using a transgenic human pluripotent stem cell (hPSC)-line that contains an inducible Neurogenin-2 (NGN2) expression cassette. The NGN2 hPSC line was first differentiated to neural crest cell (NCC) progenitors, then incorporated into a cytocompatible gelatin methacryloyl-based bioink for 3D bioprinting. Upregulated NGN2 expression in the bioprinted NCCs resulted in induced SN (iSN) populations that exhibited specific cell markers, with 3D analysis revealing widespread neurite outgrowth through the scaffold volume. Calcium imaging demonstrated functional activity of iSNs, including membrane excitability properties and voltage-gated sodium channel (NaV) activity. This efficient approach to generate 3D bioprinted iSN structures streamlines the development of neural tissue models, useful for the study of neurodevelopment and disease states and offering translational potential.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors , Bioprinting , Nerve Tissue Proteins , Printing, Three-Dimensional , Sensory Receptor Cells , Tissue Scaffolds , Humans , Bioprinting/methods , Basic Helix-Loop-Helix Transcription Factors/metabolism , Basic Helix-Loop-Helix Transcription Factors/genetics , Sensory Receptor Cells/metabolism , Sensory Receptor Cells/cytology , Nerve Tissue Proteins/metabolism , Tissue Scaffolds/chemistry , Cell Line , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Cell Differentiation , Tissue Engineering/methods , Gelatin/chemistry , Neural Crest/cytology , Neural Crest/metabolism
19.
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
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL