Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.889
Filter
Add more filters

Publication year range
1.
Cell ; 187(3): 692-711.e26, 2024 Feb 01.
Article in English | MEDLINE | ID: mdl-38262408

ABSTRACT

Transcription factors (TFs) can define distinct cellular identities despite nearly identical DNA-binding specificities. One mechanism for achieving regulatory specificity is DNA-guided TF cooperativity. Although in vitro studies suggest that it may be common, examples of such cooperativity remain scarce in cellular contexts. Here, we demonstrate how "Coordinator," a long DNA motif composed of common motifs bound by many basic helix-loop-helix (bHLH) and homeodomain (HD) TFs, uniquely defines the regulatory regions of embryonic face and limb mesenchyme. Coordinator guides cooperative and selective binding between the bHLH family mesenchymal regulator TWIST1 and a collective of HD factors associated with regional identities in the face and limb. TWIST1 is required for HD binding and open chromatin at Coordinator sites, whereas HD factors stabilize TWIST1 occupancy at Coordinator and titrate it away from HD-independent sites. This cooperativity results in the shared regulation of genes involved in cell-type and positional identities and ultimately shapes facial morphology and evolution.


Subject(s)
DNA-Binding Proteins , Embryonic Development , Transcription Factors , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Binding Sites , DNA/metabolism , DNA-Binding Proteins/metabolism , Gene Expression Regulation , Mesoderm/metabolism , Transcription Factors/metabolism , Humans , Animals , Mice , Extremities/growth & development
2.
Cell ; 186(20): 4454-4471.e19, 2023 09 28.
Article in English | MEDLINE | ID: mdl-37703875

ABSTRACT

Macrophages are heterogeneous and play critical roles in development and disease, but their diversity, function, and specification remain inadequately understood during human development. We generated a single-cell RNA sequencing map of the dynamics of human macrophage specification from PCW 4-26 across 19 tissues. We identified a microglia-like population and a proangiogenic population in 15 macrophage subtypes. Microglia-like cells, molecularly and morphologically similar to microglia in the CNS, are present in the fetal epidermis, testicle, and heart. They are the major immune population in the early epidermis, exhibit a polarized distribution along the dorsal-lateral-ventral axis, and interact with neural crest cells, modulating their differentiation along the melanocyte lineage. Through spatial and differentiation trajectory analysis, we also showed that proangiogenic macrophages are perivascular across fetal organs and likely yolk-sac-derived as microglia. Our study provides a comprehensive map of the heterogeneity and developmental dynamics of human macrophages and unravels their diverse functions during development.


Subject(s)
Macrophages , Humans , Cell Differentiation , Cell Lineage , Macrophages/cytology , Microglia , Organ Specificity
3.
Cell ; 179(1): 74-89.e10, 2019 09 19.
Article in English | MEDLINE | ID: mdl-31495570

ABSTRACT

During neural tube closure and spinal cord development, many cells die in both the central and peripheral nervous systems (CNS and PNS, respectively). However, myeloid-derived professional phagocytes have not yet colonized the trunk region during early neurogenesis. How apoptotic cells are removed from this region during these stages remains largely unknown. Using live imaging in zebrafish, we demonstrate that neural crest cells (NCCs) respond rapidly to dying cells and phagocytose cellular debris around the neural tube. Additionally, NCCs have the ability to enter the CNS through motor exit point transition zones and clear debris in the spinal cord. Surprisingly, NCCs phagocytosis mechanistically resembles macrophage phagocytosis and their recruitment toward cellular debris is mediated by interleukin-1ß. Taken together, our results reveal a role for NCCs in phagocytosis of debris in the developing nervous system before the presence of professional phagocytes.


Subject(s)
Cell Movement/physiology , Neural Crest/physiology , Neurogenesis/physiology , Peripheral Nervous System/growth & development , Phagocytosis/physiology , Spinal Cord/growth & development , Animals , Animals, Genetically Modified , Apoptosis/physiology , Cell Differentiation/physiology , Interleukin-1beta/metabolism , Phagocytes/physiology , Phagosomes/physiology , Zebrafish/embryology
4.
Cell ; 178(6): 1421-1436.e24, 2019 09 05.
Article in English | MEDLINE | ID: mdl-31491386

ABSTRACT

The developmental disorder Floating-Harbor syndrome (FHS) is caused by heterozygous truncating mutations in SRCAP, a gene encoding a chromatin remodeler mediating incorporation of histone variant H2A.Z. Here, we demonstrate that FHS-associated mutations result in loss of SRCAP nuclear localization, alter neural crest gene programs in human in vitro models and Xenopus embryos, and cause craniofacial defects. These defects are mediated by one of two H2A.Z subtypes, H2A.Z.2, whose knockdown mimics and whose overexpression rescues the FHS phenotype. Selective rescue by H2A.Z.2 is conferred by one of the three amino acid differences between the H2A.Z subtypes, S38/T38. We further show that H2A.Z.1 and H2A.Z.2 genomic occupancy patterns are qualitatively similar, but quantitatively distinct, and H2A.Z.2 incorporation at AT-rich enhancers and expression of their associated genes are both sensitized to SRCAP truncations. Altogether, our results illuminate the mechanism underlying a human syndrome and uncover selective functions of H2A.Z subtypes during development.


Subject(s)
Abnormalities, Multiple/genetics , Chromatin Assembly and Disassembly , Chromatin/metabolism , Craniofacial Abnormalities/genetics , Growth Disorders/genetics , Heart Septal Defects, Ventricular/genetics , Histones/genetics , Adenosine Triphosphatases/genetics , Amino Acid Substitution , Animals , Embryonic Stem Cells , HEK293 Cells , Humans , Mutation , Xenopus laevis
5.
Annu Rev Genet ; 55: 349-376, 2021 11 23.
Article in English | MEDLINE | ID: mdl-34546797

ABSTRACT

Neural crest stem/progenitor cells arise early during vertebrate embryogenesis at the border of the forming central nervous system. They subsequently migrate throughout the body, eventually differentiating into diverse cell types ranging from neurons and glia of the peripheral nervous system to bones of the face, portions of the heart, and pigmentation of the skin. Along the body axis, the neural crest is heterogeneous, with different subpopulations arising in the head, neck, trunk, and tail regions, each characterized by distinct migratory patterns and developmental potential. Modern genomic approaches like single-cell RNA- and ATAC-sequencing (seq) have greatly enhanced our understanding of cell lineage trajectories and gene regulatory circuitry underlying the developmental progression of neural crest cells. Here, we discuss how genomic approaches have provided new insights into old questions in neural crest biology by elucidating transcriptional and posttranscriptional mechanisms that govern neural crest formation and the establishment of axial level identity.


Subject(s)
Neural Crest , Neurons , Cell Differentiation/genetics , Cell Lineage/genetics , Cell Movement/genetics , Embryonic Development , Gene Expression Regulation, Developmental/genetics , Genomics , Neurons/metabolism
6.
Genes Dev ; 34(23-24): 1735-1752, 2020 12 01.
Article in English | MEDLINE | ID: mdl-33184218

ABSTRACT

FGFs are key developmental regulators that engage a signal transduction cascade through receptor tyrosine kinases, prominently engaging ERK1/2 but also other pathways. However, it remains unknown whether all FGF activities depend on this canonical signal transduction cascade. To address this question, we generated allelic series of knock-in Fgfr1 and Fgfr2 mouse strains, carrying point mutations that disrupt binding of signaling effectors, and a kinase dead allele of Fgfr2 that broadly phenocopies the null mutant. When interrogated in cranial neural crest cells, we identified discrete functions for signaling pathways in specific craniofacial contexts, but point mutations, even when combined, failed to recapitulate the single or double null mutant phenotypes. Furthermore, the signaling mutations abrogated established FGF-induced signal transduction pathways, yet FGF functions such as cell-matrix and cell-cell adhesion remained unaffected, though these activities did require FGFR kinase activity. Our studies establish combinatorial roles of Fgfr1 and Fgfr2 in development and uncouple novel FGFR kinase-dependent cell adhesion properties from canonical intracellular signaling.


Subject(s)
Fibroblast Growth Factors/physiology , Gene Expression Regulation, Developmental/genetics , Signal Transduction/genetics , Animals , Cell Adhesion/genetics , Cell Death/genetics , Cells, Cultured , Mice , Mutation , Neural Crest/cytology , Protein Kinases/metabolism , Receptor, Fibroblast Growth Factor, Type 1/genetics , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Receptor, Fibroblast Growth Factor, Type 2/genetics , Receptor, Fibroblast Growth Factor, Type 2/metabolism , Receptors, Fibroblast Growth Factor/genetics , Receptors, Fibroblast Growth Factor/metabolism
7.
EMBO J ; 42(1): e111139, 2023 01 04.
Article in English | MEDLINE | ID: mdl-36382711

ABSTRACT

Hirschsprung disease (HSCR), one of several neurocristopathies in children, is characterized by nerve loss in the large intestine and is mainly treated by surgery, which causes severe complications. Enteric neural crest-derived cell (ENCC) transplantation is a potential therapeutic strategy; however, so far with poor efficacy. Here, we assessed whether and how fecal microbiota transplantation (FMT) could improve ENCC transplantation in a rat model of hypoganglionosis; a condition similar to HSCR, with less intestinal innervation. We found that the hypoganglionosis intestinal microenvironment negatively influenced the ENCC functional phenotype in vitro and in vivo. Combining 16S rDNA sequencing and targeted mass spectrometry revealed microbial dysbiosis and reduced short-chain fatty acid (SCFA) production in the hypoganglionic gut. FMT increased the abundance of Bacteroides and Clostridium, SCFA production, and improved outcomes following ENCC transplantation. SCFAs alone stimulated ENCC proliferation, migration, and supported ENCC transplantation. Transcriptome-wide mRNA sequencing identified MAPK signaling as the top differentially regulated pathway in response to SCFA exposure, and inhibition of MEK1/2 signaling abrogated the SCFA-mediated effects on ENCC. This study demonstrates that FMT improves cell therapy for hypoganglionosis via short-chain fatty acid metabolism-induced MEK1/2 signaling.


Subject(s)
Fecal Microbiota Transplantation , Hirschsprung Disease , Rats , Animals , Hirschsprung Disease/therapy , Hirschsprung Disease/genetics , Hirschsprung Disease/metabolism , Signal Transduction , Fatty Acids, Volatile/metabolism , Cell- and Tissue-Based Therapy
8.
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
9.
Development ; 151(9)2024 May 01.
Article in English | MEDLINE | ID: mdl-38619396

ABSTRACT

Piezo1 and Piezo2 are recently reported mechanosensory ion channels that transduce mechanical stimuli from the environment into intracellular biochemical signals in various tissues and organ systems. Here, we show that Piezo1 and Piezo2 display a robust expression during jawbone development. Deletion of Piezo1 in neural crest cells causes jawbone malformations in a small but significant number of mice. We further demonstrate that disruption of Piezo1 and Piezo2 in neural crest cells causes more striking defects in jawbone development than any single knockout, suggesting essential but partially redundant roles of Piezo1 and Piezo2. In addition, we observe defects in other neural crest derivatives such as malformation of the vascular smooth muscle in double knockout mice. Moreover, TUNEL examinations reveal excessive cell death in osteogenic cells of the maxillary and mandibular arches of the double knockout mice, suggesting that Piezo1 and Piezo2 together regulate cell survival during jawbone development. We further demonstrate that Yoda1, a Piezo1 agonist, promotes mineralization in the mandibular arches. Altogether, these data firmly establish that Piezo channels play important roles in regulating jawbone formation and maintenance.


Subject(s)
Ion Channels , Jaw , Neural Crest , Animals , Mice , Gene Expression Regulation, Developmental , Ion Channels/metabolism , Ion Channels/genetics , Jaw/embryology , Jaw/metabolism , Mandible/embryology , Mandible/metabolism , Mice, Knockout , Neural Crest/metabolism , Osteogenesis/genetics , Pyrazines , Thiadiazoles
10.
Development ; 151(6)2024 03 15.
Article in English | MEDLINE | ID: mdl-38512806

ABSTRACT

The mechanistic target of rapamycin (mTOR) coordinates metabolism and cell growth with environmental inputs. mTOR forms two functional complexes: mTORC1 and mTORC2. Proper development requires both complexes but mTORC1 has unique roles in numerous cellular processes, including cell growth, survival and autophagy. Here, we investigate the function of mTORC1 in craniofacial development. We created a zebrafish raptor mutant via CRISPR/Cas9, to specifically disrupt mTORC1. The entire craniofacial skeleton and eyes were reduced in size in mutants; however, overall body length and developmental timing were not affected. The craniofacial phenotype associates with decreased chondrocyte size and increased neural crest cell death. We found that autophagy is elevated in raptor mutants. Chemical inhibition of autophagy reduced cell death and improved craniofacial phenotypes in raptor mutants. Genetic inhibition of autophagy, via mutation of the autophagy gene atg7, improved facial phenotypes in atg7;raptor double mutants, relative to raptor single mutants. We conclude that finely regulated levels of autophagy, via mTORC1, are crucial for craniofacial development.


Subject(s)
Neural Crest , Zebrafish , Animals , Mechanistic Target of Rapamycin Complex 1/metabolism , Zebrafish/genetics , Zebrafish/metabolism , Neural Crest/metabolism , Signal Transduction/genetics , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism , Regulatory-Associated Protein of mTOR/genetics , Regulatory-Associated Protein of mTOR/metabolism , Autophagy/genetics , Cell Death , Mutation/genetics
11.
Development ; 151(3)2024 Feb 01.
Article in English | MEDLINE | ID: mdl-38345329

ABSTRACT

The cranial sutures are proposed to be a stem cell niche, harbouring skeletal stem cells that are directly involved in development, homeostasis and healing. Like the craniofacial bones, the sutures are formed from both mesoderm and neural crest. During cranial bone repair, neural crest cells have been proposed to be key players; however, neural crest contributions to adult sutures are not well defined, and the relative importance of suture proximity is unclear. Here, we use genetic approaches to re-examine the neural crest-mesoderm boundaries in the adult mouse skull. These are combined with calvarial wounding experiments suggesting that suture proximity improves the efficiency of cranial repair. Furthermore, we demonstrate that Gli1+ and Axin2+ skeletal stem cells are present in all calvarial sutures examined. We propose that the position of the defect determines the availability of neural crest-derived progenitors, which appear to be a key element in the repair of calvarial defects.


Subject(s)
Cranial Sutures , Skull , Mice , Animals , Stem Cells , Neural Crest , Mesoderm
12.
Development ; 151(2)2024 Jan 15.
Article in English | MEDLINE | ID: mdl-38063851

ABSTRACT

Cornelia de Lange syndrome (CdLS) is a congenital disorder featuring facial dysmorphism, postnatal growth deficits, cognitive disability and upper limb abnormalities. CdLS is genetically heterogeneous, with cases arising from mutation of BRD4, a bromodomain protein that binds and reads acetylated histones. In this study, we have modeled CdLS facial pathology through mouse neural crest cell (NCC)-specific mutation of BRD4 to characterize cellular and molecular function in craniofacial development. Mice with BRD4 NCC loss of function died at birth with severe facial hypoplasia, cleft palate, mid-facial clefting and exencephaly. Following migration, BRD4 mutant NCCs initiated RUNX2 expression for differentiation to osteoblast lineages but failed to induce downstream RUNX2 targets required for lineage commitment. BRD4 bound to active enhancers to regulate expression of osteogenic transcription factors and extracellular matrix components integral for bone formation. RUNX2 physically interacts with a C-terminal domain in the long isoform of BRD4 and can co-occupy osteogenic enhancers. This BRD4 association is required for RUNX2 recruitment and appropriate osteoblast differentiation. We conclude that BRD4 controls facial bone development through osteoblast enhancer regulation of the RUNX2 transcriptional program.


Subject(s)
De Lange Syndrome , Transcription Factors , Animals , Mice , Cell Cycle Proteins/genetics , Cell Differentiation , Core Binding Factor Alpha 1 Subunit , De Lange Syndrome/genetics , Neural Crest/metabolism , Nuclear Proteins/metabolism , Osteoblasts/metabolism , Osteogenesis , Transcription Factors/metabolism
13.
Development ; 151(1)2024 Jan 01.
Article in English | MEDLINE | ID: mdl-38063857

ABSTRACT

Cranial neural crest development is governed by positional gene regulatory networks (GRNs). Fine-tuning of the GRN components underlies facial shape variation, yet how those networks in the midface are connected and activated remain poorly understood. Here, we show that concerted inactivation of Tfap2a and Tfap2b in the murine neural crest, even during the late migratory phase, results in a midfacial cleft and skeletal abnormalities. Bulk and single-cell RNA-seq profiling reveal that loss of both TFAP2 family members dysregulates numerous midface GRN components involved in midface morphogenesis, patterning and differentiation. Notably, Alx1, Alx3 and Alx4 (ALX) transcript levels are reduced, whereas ChIP-seq analyses suggest TFAP2 family members directly and positively regulate ALX gene expression. Tfap2a, Tfap2b and ALX co-expression in midfacial neural crest cells of both mouse and zebrafish implies conservation of this regulatory axis across vertebrates. Consistent with this notion, tfap2a zebrafish mutants present with abnormal alx3 expression patterns, Tfap2a binds ALX loci and tfap2a-alx3 genetic interactions are observed. Together, these data demonstrate TFAP2 paralogs regulate vertebrate midfacial development in part by activating expression of ALX transcription factor genes.


Subject(s)
Zebrafish Proteins , Zebrafish , Animals , Mice , Zebrafish/genetics , Zebrafish/metabolism , Zebrafish Proteins/genetics , Cell Differentiation/genetics , Transcription Factor AP-2/genetics , Transcription Factor AP-2/metabolism , Genes, Homeobox , Neural Crest , Gene Expression Regulation, Developmental
14.
Development ; 151(13)2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38884356

ABSTRACT

Neural crest cells are a stem cell population unique to vertebrate embryos that retains broad multi-germ layer developmental potential through neurulation. Much remains to be learned about the genetic and epigenetic mechanisms that control the potency of neural crest cells. Here, we examine the role that epigenetic readers of the BET (bromodomain and extra terminal) family play in controlling the potential of pluripotent blastula and neural crest cells. We find that inhibiting BET activity leads to loss of pluripotency at blastula stages and a loss of neural crest at neurula stages. We compare the effects of HDAC (an eraser of acetylation marks) and BET (a reader of acetylation) inhibition and find that they lead to similar cellular outcomes through distinct effects on the transcriptome. Interestingly, loss of BET activity in cells undergoing lineage restriction is coupled to increased expression of genes linked to pluripotency and prolongs the competence of initially pluripotent cells to transit to a neural progenitor state. Together these findings advance our understanding of the epigenetic control of pluripotency and the formation of the vertebrate neural crest.


Subject(s)
Neural Crest , Animals , Neural Crest/cytology , Neural Crest/metabolism , Epigenesis, Genetic , Gene Expression Regulation, Developmental , Xenopus Proteins/metabolism , Xenopus Proteins/genetics , Xenopus laevis/embryology , Blastula/metabolism , Blastula/cytology , Cell Differentiation , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Transcriptome/genetics
15.
Development ; 151(14)2024 Jul 15.
Article in English | MEDLINE | ID: mdl-38940470

ABSTRACT

SoxB1 transcription factors (Sox2/3) are well known for their role in early neural fate specification in the embryo, but little is known about functional roles for SoxB1 factors in non-neural ectodermal cell types, such as the neural plate border (NPB). Using Xenopus laevis, we set out to determine whether SoxB1 transcription factors have a regulatory function in NPB formation. Here, we show that SoxB1 factors are necessary for NPB formation, and that prolonged SoxB1 factor activity blocks the transition from a NPB to a neural crest state. Using ChIP-seq, we demonstrate that Sox3 is enriched upstream of NPB genes in early NPB cells and in blastula stem cells. Depletion of SoxB1 factors in blastula stem cells results in downregulation of NPB genes. Finally, we identify Pou5f3 factors as potential Sox3 partners in regulating the formation of the NPB and show that their combined activity is needed for normal NPB gene expression. Together, these data identify a role for SoxB1 factors in the establishment and maintenance of the NPB, in part through partnership with Pou5f3 factors.


Subject(s)
Gene Expression Regulation, Developmental , Neural Crest , Neural Plate , SOXB1 Transcription Factors , Xenopus Proteins , Xenopus laevis , Animals , Neural Plate/metabolism , Neural Plate/embryology , SOXB1 Transcription Factors/metabolism , SOXB1 Transcription Factors/genetics , Xenopus Proteins/metabolism , Xenopus Proteins/genetics , Neural Crest/metabolism , Neural Crest/cytology , Blastula/metabolism , Embryo, Nonmammalian/metabolism
16.
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
17.
Annu Rev Genet ; 53: 505-530, 2019 12 03.
Article in English | MEDLINE | ID: mdl-31509458

ABSTRACT

Vertebrate pigment patterns are diverse and fascinating adult traits that allow animals to recognize conspecifics, attract mates, and avoid predators. Pigment patterns in fish are among the most amenable traits for studying the cellular basis of adult form, as the cells that produce diverse patterns are readily visible in the skin during development. The genetic basis of pigment pattern development has been most studied in the zebrafish, Danio rerio. Zebrafish adults have alternating dark and light horizontal stripes, resulting from the precise arrangement of three main classes of pigment cells: black melanophores, yellow xanthophores, and iridescent iridophores. The coordination of adult pigment cell lineage specification and differentiation with specific cellular interactions and morphogenetic behaviors is necessary for stripe development. Besides providing a nice example of pattern formation responsible for an adult trait of zebrafish, stripe-forming mechanisms also provide a conceptual framework for posing testable hypotheses about pattern diversification more broadly. Here, we summarize what is known about lineages and molecular interactions required for pattern formation in zebrafish, we review some of what is known about pattern diversification in Danio, and we speculate on how patterns in more distant teleosts may have evolved to produce a stunningly diverse array of patterns in nature.


Subject(s)
Pigmentation/physiology , Zebrafish/physiology , Animals , Biological Evolution , Cell Lineage , Melanophores/physiology , Neural Crest , Paracrine Communication , Zebrafish/genetics , Zebrafish/growth & development , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
18.
Proc Natl Acad Sci U S A ; 121(3): e2317668121, 2024 Jan 16.
Article in English | MEDLINE | ID: mdl-38194455

ABSTRACT

Orofacial clefts of the lip and palate are widely recognized to result from complex gene-environment interactions, but inadequate understanding of environmental risk factors has stymied development of prevention strategies. We interrogated the role of DNA methylation, an environmentally malleable epigenetic mechanism, in orofacial development. Expression of the key DNA methyltransferase enzyme DNMT1 was detected throughout palate morphogenesis in the epithelium and underlying cranial neural crest cell (cNCC) mesenchyme, a highly proliferative multipotent stem cell population that forms orofacial connective tissue. Genetic and pharmacologic manipulations of DNMT activity were then applied to define the tissue- and timing-dependent requirement of DNA methylation in orofacial development. cNCC-specific Dnmt1 inactivation targeting initial palate outgrowth resulted in OFCs, while later targeting during palatal shelf elevation and elongation did not. Conditional Dnmt1 deletion reduced cNCC proliferation and subsequent differentiation trajectory, resulting in attenuated outgrowth of the palatal shelves and altered development of cNCC-derived skeletal elements. Finally, we found that the cellular mechanisms of cleft pathogenesis observed in vivo can be recapitulated by pharmacologically reducing DNA methylation in multipotent cNCCs cultured in vitro. These findings demonstrate that DNA methylation is a crucial epigenetic regulator of cNCC biology, define a critical period of development in which its disruption directly causes OFCs, and provide opportunities to identify environmental influences that contribute to OFC risk.


Subject(s)
Cleft Lip , Cleft Palate , Animals , Mice , Cleft Lip/genetics , DNA Methylation , Cleft Palate/genetics , Neural Crest , DNA Modification Methylases , Cell Proliferation
19.
Proc Natl Acad Sci U S A ; 121(19): e2311685121, 2024 May 07.
Article in English | MEDLINE | ID: mdl-38683994

ABSTRACT

Neural crest cells exemplify cellular diversification from a multipotent progenitor population. However, the full sequence of early molecular choices orchestrating the emergence of neural crest heterogeneity from the embryonic ectoderm remains elusive. Gene-regulatory-networks (GRN) govern early development and cell specification toward definitive neural crest. Here, we combine ultradense single-cell transcriptomes with machine-learning and large-scale transcriptomic and epigenomic experimental validation of selected trajectories, to provide the general principles and highlight specific features of the GRN underlying neural crest fate diversification from induction to early migration stages using Xenopus frog embryos as a model. During gastrulation, a transient neural border zone state precedes the choice between neural crest and placodes which includes multiple converging gene programs. During neurulation, transcription factor connectome, and bifurcation analyses demonstrate the early emergence of neural crest fates at the neural plate stage, alongside an unbiased multipotent-like lineage persisting until epithelial-mesenchymal transition stage. We also decipher circuits driving cranial and vagal neural crest formation and provide a broadly applicable high-throughput validation strategy for investigating single-cell transcriptomes in vertebrate GRNs in development, evolution, and disease.


Subject(s)
Neural Crest , Single-Cell Analysis , Xenopus laevis , Animals , Neural Crest/cytology , Neural Crest/metabolism , Single-Cell Analysis/methods , Xenopus laevis/embryology , Gene Expression Regulation, Developmental , Cell Movement , Gene Regulatory Networks , Transcriptome , Gastrulation , Neural Plate/metabolism , Neural Plate/embryology , Neural Plate/cytology , Epithelial-Mesenchymal Transition/genetics , Embryo, Nonmammalian/metabolism , Embryo, Nonmammalian/cytology , Neurulation/genetics , Neurulation/physiology , Cell Differentiation
20.
EMBO J ; 41(17): e108780, 2022 09 01.
Article in English | MEDLINE | ID: mdl-35815410

ABSTRACT

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.


Subject(s)
Neural Crest , Schwann Cells , Cell Differentiation/physiology , Neurogenesis/physiology , Peripheral Nerves , Schwann Cells/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL