Fact-checked by Grok 2 weeks ago

Neural crest

The neural crest is a unique, transient population of multipotent cells in embryos that originates at the border between the and non-, undergoing epithelial-to-mesenchymal transition to migrate extensively and differentiate into diverse derivatives such as peripheral neurons, , melanocytes, craniofacial and , and adrenal chromaffin cells. This embryonic structure, often dubbed the "fourth germ layer" due to its pivotal role in development, emerges during around the third week of in humans, specifically between days 21 and 28 post-fertilization. Neural crest cells (NCCs) are specified early in at the border through interactions involving signaling molecules like , Wnt, and FGF, which activate a conserved including transcription factors such as , , and Snail2. Their development proceeds along a rostrocaudal axis, with cephalic NCCs contributing to head and neck structures like the and cranial ganglia, cardiac NCCs forming elements of the heart's outflow tract, NCCs giving rise to dorsal root ganglia and melanocytes, and vagal/sacral NCCs populating the . Migration is tightly regulated by extracellular cues, including ephrins, semaphorins, and components, allowing NCCs to delaminate from the dorsal and follow stereotypical pathways while avoiding barriers like the somitic sclerotome. Evolutionarily, the neural crest represents a hallmark of , appearing over 500 million years ago and enabling the formation of complex features like the vertebrate head and , transforming the ancestral . This multipotency, akin to behavior with limited self-renewal, underscores the neural crest's role in and highlights its clinical relevance in neurocristopathies—disorders such as Hirschsprung disease, , and neuroblastomas arising from NCC dysfunction or malignancy.

Overview

Definition and characteristics

The neural crest is a transient, multipotent cell population that arises at the dorsal aspect of the during early embryogenesis. These cells originate from the and represent a unique developmental entity capable of giving rise to a wide array of cell types across multiple tissue systems. Unlike traditional germ layers, the neural crest's versatility in contributing to both neural and non-neural derivatives underscores its foundational role in formation. Key characteristics of neural crest cells include their ability to undergo epithelial-to-mesenchymal transition (), enabling delamination from the epithelium, followed by extensive long-distance through the embryo and subsequent differentiation into diverse lineages such as neurons, , melanocytes, and craniofacial . Morphologically, premigratory neural crest cells form part of the pseudostratified epithelium at the border, where they express specific transcription factors including and FoxD3, which are essential for maintaining their multipotency and initiating their developmental program. Due to these extensive contributions that extend beyond the scope of the , , and —the three primary s—the neural crest is often classified as a "fourth germ layer" in development. This designation highlights its evolutionary significance as a innovation, conserved across all species but absent in , thereby distinguishing from other chordates.

Embryonic origin and timing

The neural crest originates at the border, a transitional zone between the prospective neural ectoderm and the non-neural surface , during the stages of embryogenesis. This border region emerges as the forms, positioning the neural crest dorsally along the developing neural axis. The formation of the neural crest is closely linked to the dynamic of the , where convergent extension movements narrow and elongate the tissue, elevating the lateral edges into neural folds that enclose the . These processes integrate signals from adjacent tissues, including the underlying and overlying surface , which help stabilize the border domain and refine its identity before . Timing of neural crest formation varies across vertebrate species but generally aligns with neural tube closure. In mice, specification of the neural crest begins around embryonic day 8.5 (E8.5), with initial cell emergence from the dorsal neural tube occurring between E8.5 and E9.5, and full delamination extending to E10.5 along the trunk axis. In humans, this process takes place during the third to fourth weeks of gestation, coinciding with early somitogenesis and neural tube formation. In chick embryos, neural crest progenitors are specified at Hamburger-Hamilton (HH) stages 8-9, with overt formation and initial migration evident by HH stage 10. Across species, neural crest formation exhibits axial variations along the anterior-posterior axis, progressing in a wave-like manner from cranial to caudal regions. Anterior (cranial) neural crest emerges first, contributing to head structures, while trunk and posterior (vagal and sacral) populations follow sequentially, reflecting the spatiotemporal progression of and formation. This patterned emergence ensures coordinated development, with interactions between the forming , surface , and paraxial modulating the timing and extent of crest production at each level.

Developmental Biology

Induction and specification

The induction of the neural crest begins during in vertebrate embryos, where interactions between the and underlying establish the neural plate border, a transient zone that gives rise to neural crest precursors. Key inductive signals emanate from the dorsal and , including bone morphogenetic proteins (), Wnts, and fibroblast growth factors (FGFs), which promote the formation of this border domain. Specifically, signaling, initially inhibited by antagonists like Noggin during early to allow neural induction, is later activated at neurula stages to maintain neural crest competence, often in synergy with canonical Wnt/β-catenin and FGF pathways that regulate downstream effectors. These signals induce border specifiers that suppress neural genes such as , preventing full neural commitment while priming cells for neural crest fate. The neural plate border is specified by a set of transcription factors that interpret these inductive cues, including homeobox genes like Msx1 and Dlx family members, as well as paired-box genes Pax3 and Pax7. These border specifiers are expressed early in gastrulation and define the transitional domain between presumptive neural and epidermal tissues, integrating BMP, Wnt, and FGF inputs to restrict alternative fates. Subsequent commitment to the neural crest lineage involves a core set of neural crest specifiers, such as the SRY-related HMG-box factors Sox9 and Sox10, the forkhead factor FoxD3, and the basic helix-loop-helix protein Twist1. These factors form a hierarchical gene regulatory network (GRN) that stabilizes neural crest identity, with upstream border specifiers activating the core module during late gastrulation to early neurulation. Downstream effector genes, including the zinc-finger transcription factor Snail2 (also known as Slug) and the GTPase RhoB, are then upregulated to prepare cells for epithelial-to-mesenchymal transition (EMT), though full delamination occurs later. Classic experimental models, such as grafting assays, have confirmed the necessity of these inductive signals and components for neural crest formation. In these chimeras, transplantation of presumptive neural crest regions from quail donors into hosts at gastrula stages demonstrates that exposure to /Wnt/FGF-rich environments is required for border specification and specifier expression, with ablation of dorsal signals abolishing neural crest markers like Sox10. More recent advances using single-cell sequencing (scRNA-seq) have revealed the dynamic nature of specifier expression during , showing transitional states from border to premigratory neural crest cells with heterogeneous activation of Sox9, Sox10, and FoxD3 in species like , , and , highlighting probabilistic fate decisions within the . Epigenetic mechanisms further refine neural crest specification through Polycomb group (PcG) proteins, which establish bivalent domains at key loci. In cranial neural crest cells, the PcG component deposits marks alongside activating H3K4me2, poising positional identity genes (e.g., those directing craniofacial fates) for rapid activation by local signals post-specification. This bivalency, observed in over 80% of relevant promoters at embryonic day 10.5 in mouse, maintains multipotency within the GRN and prevents premature differentiation, ensuring adaptability to environmental cues during subsequent development.

Migration and delamination

Neural crest cells undergo delamination from the dorsal through an epithelial-to-mesenchymal transition (), a that enables their and subsequent . During , neural crest cells downregulate epithelial such as N-cadherin (CDH2), facilitating a shift toward mesenchymal characteristics and to fibronectin-rich substrates. This cadherin switch is complemented by the upregulation of cadherin-11 (CDH11), which supports cell survival and motility post-delamination. activation, particularly matrix metalloproteinases (MMPs) like MMP2 and MMP9, plays a critical role by degrading basement membrane components, including and IV, to create channels; for instance, MMP9 targets CDH2 and to promote in chick cranial neural crest cells. Following , neural crest cells along distinct pathways, including dorsal routes toward the skin and , and ventral paths to the pharyngeal arches and gut. Guidance during is provided by the (ECM), with and serving as permissive substrates that interact with such as α5β1 to direct cell movement. like SDF-1 () and its receptor further orient , particularly in cardiac neural crest cells, where signaling from placodal cells and the influences trajectories in and embryos. Neural crest cells also secrete to remodel the ECM ahead of their advance, enhancing in permissive corridors. Migration occurs in both collective and individual modes, with trunk neural crest cells often forming chainlike arrays through rostral somites via filopodial contacts and N-cadherin-mediated adhesion, while individual cells exhibit biased random walks but show increased directionality in groups. Inhibition zones, such as those in caudal somites, restrict entry through repulsive signals like ephrin-B1 and semaphorins, confining cells to rostral pathways; non-canonical Wnt/ signaling supports chain formation but does not directly inhibit in rostral regions. Regulatory factors include signaling, where neuregulin-1 (NRG1) from the dorsal aorta times trunk neural crest emigration in embryos. Semaphorins (e.g., Sema3A, Sema3F) and ephrins (e.g., ephrin-B1) guide by repulsion at boundaries, as seen in cephalic and neural crest in and models. Recent studies using live imaging have revealed collective delamination waves, where approximately 20-30% of cranial cells in embryos exit via rather than pure , driven by mechanical forces sensed through channels under tissue tension. This mechanism, observed in time-lapse sequences over 130 minutes, transitions cells to a mesenchymal state post-exit, highlighting the role of mechanical cues in . Experimental evidence from time-lapse microscopy in and demonstrates that confinement by components like versican promotes collective migration in streams, with optimal cluster sizes correlating to pathway width for enhanced directionality. In trunk neural crest, 3D confocal imaging shows chain formation along the anterior-posterior axis, while grafts reveal disrupted upon versican knockdown.

Derivatives and Lineages

Neural derivatives

The neural crest gives rise to a diverse array of neural cell types that form critical components of the peripheral nervous system (PNS), including sensory and autonomic neurons as well as glial cells. These derivatives arise from multipotent neural crest cells that undergo specification and differentiation influenced by intrinsic genetic programs and extrinsic environmental signals. In the trunk region, neural crest cells contribute to the formation of dorsal root ganglia (DRG), which house sensory neurons responsible for transmitting somatosensory information from the body to the . Similarly, cranial neural crest cells populate the trigeminal ganglia, providing sensory innervation to the face and oral cavity. Autonomic neurons also originate from neural crest progenitors, with sympathetic neurons deriving primarily from trunk-level crest cells that migrate ventrally to form paravertebral and prevertebral ganglia, enabling responses such as and increased through noradrenergic signaling. Parasympathetic neurons, in contrast, emerge from vagal and sacral neural crest populations, contributing to ganglia that regulate visceral functions like and glandular via transmission. Schwann cells, the myelinating glia of the PNS, envelop peripheral axons to facilitate rapid nerve conduction and provide trophic support; these cells trace their lineage to neural crest-derived precursors, which arise from bipotent glia-neuron progenitors during early development. The (ENS), often termed the "second brain," forms an extensive network of neurons and glia within the , primarily colonized by vagal neural crest cells that migrate rostrocaudally along the gut, with additional contributions from sacral crest cells to the . Differentiation of these neural lineages is guided by key signaling molecules, including neurotrophins such as (NGF) and (BDNF), which promote neuronal survival and outgrowth—NGF supporting sympathetic and maintenance, while BDNF influences specification from pluripotent crest cells. Notch signaling plays a pivotal role in promoting glial fates, such as differentiation, by inhibiting neuronal differentiation in bipotent progenitors through mechanisms. Recent studies have identified neural crest stem cells (NCSCs) persisting in the adult PNS, particularly in the enteric ganglia and peripheral nerves, where they retain multipotency to generate neurons and , offering potential for tissue regeneration following injury. These adult NCSCs can be isolated from sites like the gut or and differentiate into functional PNS components , highlighting their therapeutic promise for repairing peripheral neuropathies.

Non-neural derivatives

The neural crest gives rise to a diverse array of non-neural cell types, primarily through ectomesenchymal lineages that contribute to connective, skeletal, endocrine, and pigmentary tissues across the body. These derivatives arise from specific regional populations of neural crest cells, which undergo epithelial-to-mesenchymal transition and migrate to distant sites before differentiating into specialized functions. Unlike neural fates, these non-neural contributions emphasize structural support, secretion, and pigmentation, highlighting the multipotency of neural crest progenitors. Melanocytes, the pigment-producing cells responsible for , , and eye coloration, originate exclusively from trunk neural crest cells. These cells migrate dorsolaterally through the developing to populate the , where they produce via melanosomes to protect against UV . Differentiation and survival of melanocyte precursors, known as melanoblasts, depend on signaling, a pathway activated by (SCF), which promotes proliferation, migration, and resistance to during their journey from the neural crest to peripheral tissues. Adrenal chromaffin cells, which form the endocrine component of the , derive from trunk neural crest progenitors that migrate ventrally to associate with the developing . These cells secrete catecholamines such as adrenaline and noradrenaline in response to stress, playing a critical role in the . Chromaffin cells share a common sympathoadrenal lineage with sympathetic neurons, diverging through differential expression of transcription factors like Hand2 and Phox2b, which suppress neuronal traits while promoting neuroendocrine differentiation; their similarity to neurons is evident in shared synthesis pathways but distinct in lacking axons. Cranial neural crest cells contribute substantially to the craniofacial , forming the and elements of the head and face through ectomesenchymal and chondrogenesis. For instance, cells populating the first differentiate into Meckel's cartilage, a transient structure that serves as a template for the and associated like the and . These progenitors undergo skeletogenic under the influence of signals such as and FGF from the pharyngeal , resulting in the membranous and endochondral bones that define facial architecture. Neural crest-derived ectomesenchyme also forms key connective tissues in the eye and teeth. In the eye, periocular neural crest cells migrate to the anterior chamber, differentiating into the , a essential for maintaining corneal transparency through fluid transport and . In dental , neural crest cells contribute to the and follicle, providing connective tissue stroma that supports odontogenesis and periodontal ligament formation. Odontoblasts, the dentin-secreting cells lining the , originate from cranial neural crest-derived ectomesenchymal cells within the . These columnar cells extend processes into the matrix, depositing collagen-rich predentin that mineralizes to form the protecting the . Their is induced by epithelial-mesenchymal interactions during , involving transcription factors like Msx1 and to initiate matrix secretion and sensory innervation integration. From the cardiac neural crest, progenitors migrate into the outflow tract and pharyngeal arches, differentiating into cells that ensheath the great vessels, including the . These cells provide structural integrity and contractile properties to the tunica media of the and pulmonary trunk, essential for cardiovascular . Their requires signals like and TGF-β to promote mesenchymal-to-smooth muscle transition, ensuring proper remodeling of the arterial system during septation. Cranial neural crest cells also contribute to the leptomeninges, specifically the and , which envelop the . These layers provide structural support, facilitate cerebrospinal fluid circulation, and contribute to the blood-brain barrier formation. Neural crest-derived meningeal fibroblasts arise from ectomesenchymal progenitors and integrate with mesodermal components to form the protective meningeal coverings. Recent studies have clarified additional non-neural contributions, including neural crest-derived mesenchymal cells in the thymic stroma. These cells, originating from third pharyngeal pouch neural crest, integrate into the thymic to support epithelial organization and T-cell maturation through production and signaling. Single-cell analyses reveal their dynamic role in thymus , distinguishing them from mesodermal stromal components.

Regional variations

The neural crest exhibits significant regional variations along the anteroposterior axis of the , with cells originating from distinct axial levels displaying differences in migratory behavior, developmental potency, and derivative contributions. These variations arise during the specification phase and are influenced by positional cues, leading to subpopulations such as cranial, cardiac, vagal, , and sacral neural crest. Foundational lineage-tracing studies in and murine models have demonstrated that neural crest cells from anterior regions contribute to a broader array of tissues compared to those from more posterior levels, reflecting an intrinsic axial identity. Cranial neural crest cells, arising from the and regions anterior to the otic vesicle, possess high ectomesenchymal potential and migrate in streams to populate the pharyngeal arches. These cells give rise to the , including cranial bones and , as well as cranial ganglia such as the trigeminal and vestibulocochlear ganglia. Unlike more posterior populations, cranial neural crest cells exhibit plasticity in morphogenesis, with environmental cues in the branchial arches guiding their differentiation into mesenchymal derivatives rather than strict fate restriction prior to migration. Cardiac neural crest cells originate from the post-otic hindbrain, specifically rhombomeres 6-8, and migrate through the pharyngeal arches to the outflow tract of the developing heart. They play a critical role in septation of the cardiac outflow tract, contributing to the division between the aorta and pulmonary trunk, and provide parasympathetic innervation to the heart via neurons in the cardiac ganglia. Ablation experiments in chick embryos have shown that these cells are essential for proper alignment of the great arteries, underscoring their specialized cardiovascular function. Vagal and sacral neural crest cells are specialized for (ENS) colonization, with vagal cells emerging from the adjacent to somites 1-7 to innervate the , and sacral cells from the caudal (somites 24-28) targeting the . Vagal neural crest cells migrate extensively along the gut axis, diversifying into neurons and that form the ENS network, while sacral cells provide a secondary contribution to the distal regions. This division reflects a transitional role between cranial and trunk populations, with vagal cells showing intermediate migratory behaviors. Trunk neural crest cells, derived from the thoracic and lumbar spinal cord levels (somites 8-23), primarily generate neural derivatives such as peripheral nervous system (PNS) neurons and glia, including dorsal root ganglia and sympathetic chain, as well as melanocytes that migrate dorsolaterally to the skin. These cells have limited skeletogenic ability compared to cranial populations, with migration pathways strictly guided by prior fate specification, such as ventromedial routes for neural fates and dorsolateral for melanoblasts. A key feature of neural crest regionalization is the potency gradient along the anteroposterior axis, where anterior (cranial) cells are multipotent, capable of mesenchymal and neural fates, while posterior (trunk and sacral) cells are more restricted to neural lineages. This gradient has been evidenced by heterotopic transplantation experiments, in which cranial neural crest cells transplanted to trunk levels can adopt skeletogenic fates, but trunk cells placed anteriorly fail to form . Hox genes further refine this regional identity by establishing axial patterning; for instance, Hoxa2 specifies cranial in pharyngeal arches, while posterior Hox clusters (e.g., Hox6-9) define trunk and sacral domains. Recent single-cell sequencing (scRNA-seq) studies have revealed heterogeneity underlying these regional differences, identifying transcriptionally distinct subpopulations at early stages. In murine embryos, scRNA-seq of delaminating neural crest cells showed cranial s biased toward mesenchymal genes (e.g., Twist1, Prrx1), while trunk s expressed neuronal markers (e.g., Neurog2), with bipotent intermediates marking fate bifurcations. further confirmed Hox-dependent clustering, highlighting how molecular heterogeneity emerges prior to migration and contributes to the observed potency gradients.

Clinical Significance

Neurocristopathies overview

Neurocristopathies are a diverse group of congenital disorders resulting from abnormalities in neural crest , , or during embryogenesis. These conditions arise because neural crest cells, which are multipotent progenitors contributing to multiple tissues, fail to properly specify, delaminate, or populate target sites, leading to defects in derivatives such as the peripheral , craniofacial structures, and melanocytes. Common themes in neurocristopathies include genetic mutations affecting key transcription factors that regulate neural crest potency and migration, such as , which is essential for maintaining multipotency and directing differentiation. For instance, mutations disrupt the proliferation, migration, and survival of neural crest cells, resulting in reduced cellular potency and impaired lineage commitment. Migration failures are particularly prevalent, often causing incomplete colonization of tissues like the (ENS). Neurocristopathies are classified into syndromic forms, which involve multi-system involvement (e.g., craniofacial, cardiac, and pigmentary anomalies), and isolated forms affecting a single system or tissue. Syndromic examples include , while isolated cases like , characterized by aganglionic bowel segments due to failed ENS migration, have a prevalence of approximately 1 in 5,000 live births. Pathophysiological mechanisms encompass dysregulation of , which prematurely eliminates neural crest cells, and incomplete leading to , such as in the ENS or craniofacial . Craniofacial dysmorphology often stems from defective neural crest contributions to skeletal and connective s, while ENS defects result in functional gastrointestinal impairments. Recent advances from 2024 to 2025 have enhanced understanding through genetic screening techniques, such as multi-omics approaches and targeted panels for neural crest genes, identifying novel variants in conditions like . models, including induced pluripotent stem cell-derived neural crest organoids, have enabled recapitulation of disease phenotypes and testing of therapeutic interventions, such as gene editing to restore migration. Diagnostic approaches typically combine imaging modalities, like MRI and for craniofacial or vascular anomalies, with using panels targeting neural crest specifier genes to confirm etiology and guide management.

Specific disorders

is a neurocristopathy characterized by , pigmentation abnormalities, and sometimes dystopia canthorum, arising from defects in neural crest-derived melanocytes and otic vesicles. Mutations in the gene, encoding a essential for neural crest cell survival and , cause type 1 Waardenburg syndrome (WS1) by disrupting melanocyte development in the skin, hair, and . Similarly, mutations, which impair the regulation of genes like MITF involved in melanocyte lineage specification, underlie type 2 (WS2) and type 4 (WS4) forms, leading to reduced neural crest progenitor proliferation and migration to pigmentary and auditory structures. These genetic alterations result in incomplete , with affecting approximately 60% of WS1 cases and 70-90% of WS2 cases due to failed inner ear melanocyte function. Hirschsprung's disease (HSCR) manifests as aganglionic from the failure of enteric neural crest cells (ENCCs) to migrate, , and differentiate into the (ENS), particularly from vagal neural crest origins. The RET proto-oncogene, a critical for ENCC guidance via GDNF signaling, accounts for up to 50% of familial and 15-35% of sporadic HSCR cases through loss-of-function variants that halt distal gut colonization. EDNRB variants, encoding the B receptor involved in ENCC and , contribute to 5% of cases, often in syndromic forms with pigmentation defects, exacerbating the absence of ganglion cells in the distal bowel and leading to functional obstruction. or digenic inheritance involving RET and EDNRB can shift ENCC fate, resulting in variable aganglionic segments from short-segment to total colonic involvement. DiGeorge syndrome (22q11.2 deletion syndrome) features conotruncal heart defects, thymic , and due to impaired cardiac neural crest cell contributions to outflow tract septation and pharyngeal pouch development. The TBX1 gene within the deleted region encodes a T-box that regulates neural crest cell migration and survival in the pharyngeal arches, with causing abnormal remodeling and thymic in approximately 60-80% of cases, with rare complete aplasia. TBX1 modulates signaling and downstream targets like FGF8, disrupting the non-autonomous interactions between neural crest and endodermal cells essential for third/fourth pharyngeal pouch formation. This leads to interrupted or in approximately 75% of patients, alongside immune deficiencies from failed T-cell maturation. Treacher Collins syndrome (TCS) involves mandibular hypoplasia, downslanting palpebral fissures, and from craniofacial skeletal malformations originating in cranial neural crest cells. Mutations in TCOF1, encoding the nucleolar protein , disrupt ribosomal biogenesis and increase neuroepithelial during neural crest induction, reducing the progenitor pool available for first and second derivatives. of TCOF1 elevates p53-mediated cell death in prefusion neural folds, leading to hypoplastic and zygoma in 90% of cases, with variable severity due to autosomal dominant . Treacle's role in transcription is critical for neural crest , and its deficiency impairs mesenchymal in facial prominences. Fetal alcohol spectrum disorder (FASD) encompasses a range of craniofacial, cardiac, and neurodevelopmental anomalies from prenatal exposure disrupting neural crest cell induction and migration. inhibits Sonic hedgehog (SHH) signaling and increases , reducing cranial neural crest cell survival and altering frontonasal and maxillary process fusion, resulting in midface and smooth in affected individuals. Cardiac defects like ventricular septal defects arise from impaired cardiac neural crest outflow tract contributions, with perturbing and Wnt pathways essential for crest cell . Exposure during to stages heightens vulnerability, with facial dysmorphology serving as a for neural crest disruption. CHARGE syndrome, caused by CHD7 mutations, presents with coloboma, heart defects, , retarded growth, genital anomalies, and ear abnormalities from widespread neural crest dysfunction. CHD7, a remodeler, regulates enhancers for genes like SEMA3 and ROBO1 involved in neural crest guidance, with loss-of-function leading to defective migration and differentiation in cranial and cardiac crest populations. Mutations disrupt ATP-dependent remodeling, impairing neural crest specification and contributing to conotruncal anomalies and malformations in 80-90% of cases. Neuroblastoma, a pediatric , arises from sympathoadrenal neural crest progenitors due to proliferative and differentiative defects, often involving MYCN that drives uncontrolled trunk neural crest expansion. High-risk cases exhibit arrested at the sympathoblast , leading to adrenal or paraspinal tumors with metastatic potential in 50% of patients under age 5. Neural crest origin is evidenced by expression of markers like PHOX2B, with genomic instability from ALK or alterations exacerbating oncogenic transformation. Emerging therapies for neurocristopathies target neural crest defects directly. CRISPR/Cas9 editing has corrected RET mutations (e.g., G731del) in patient-derived induced pluripotent stem cells (iPSCs), restoring ENCC migration and functionality in Hirschsprung models. Enteric (ENSC) transplants from human iPSCs repopulate aganglionic colon in preclinical studies, improving gut by 40-60% in HSCR models through grafted integration. These approaches hold promise for vagal and cardiac crest-related disorders, though clinical translation requires addressing engraftment efficiency and immune compatibility.

Evolution

Origins in chordates

The neural crest, a transient population of multipotent cells unique to vertebrates, is absent in non-chordate animals such as fruit flies (Drosophila melanogaster) and nematodes (Caenorhabditis elegans), where no equivalent migratory ectodermal cells with similar gene regulatory networks (GRNs) or developmental potential have been identified. This absence underscores the neural crest as a chordate-specific innovation that arose during the early evolution of vertebrates, approximately 520 million years ago. In the basal chordate group Cephalochordata, represented by amphioxus (Branchiostoma species), the neural crest first emerges in a rudimentary form as non-migratory border cells at the edges of the neural plate. These cells express a subset of vertebrate neural crest markers, including Msx, Dlx, and Zic genes, but lack the full epithelial-to-mesenchymal transition (EMT) and multipotency characteristic of vertebrate neural crest. This partial GRN in amphioxus suggests an evolutionary precursor to the neural crest, rooted in conserved signaling pathways like BMP and Wnt that pattern the neural plate border across bilaterians, though full EMT and delamination occur only in more derived chordates. Among Urochordata (tunicates), such as and Ecteinascidia turbinata, more advanced neural crest homologs appear as migratory cells originating from the borders, expressing genes like Delta-Notch, , , Pax3/7, and FoxD, which drive limited migration and differentiation into pigment cells or sensory neurons. These cells exhibit partial multipotency but not the broad developmental repertoire of neural crest, indicating an intermediate evolutionary stage. Recent phylogenetic analyses, bolstered by genomic data from 2023 onward, confirm that urochordates form the to vertebrates within the , with cephalochordates as the basal outgroup to this pairing, implying that neural crest-like features evolved after the divergence from amphioxus but before the tunicate-vertebrate split around 520 million years ago. This positioning highlights urochordates as the closest models for studying neural crest origins, with shared migratory progenitors providing insights into the transition to vertebrate innovations. The emergence of the neural crest in chordates is closely linked to the "New Head" hypothesis, where these multipotent cells enabled the evolutionary elaboration of the cranium, sensory organs, and branchial structures by contributing mesenchymal components to the head . Indirect fossil evidence from , such as Myllokunmingia and (~520 million years ago), supports this through the presence of proto-branchial arches and dermal armor, interpreted as neural crest-derived based on their mesenchyme-like composition and position. While GRNs involving and Wnt are conserved in invertebrate neural borders for ectodermal patterning, the chordate-specific integration of these with EMT regulators like and facilitated the neural crest's role in head diversification.

Role in vertebrate diversification

The neural crest played a pivotal role in the of the head, particularly through the cranial neural crest, which contributed to the formation of and sensory structures in gnathostomes (jawed s). This innovation marked a significant diversification from jawless ancestors, enabling predatory behaviors and the "new head" hypothesis, where neural crest cells provided mesenchymal contributions to the craniofacial , including branchial arches that developed into . In gnathostomes, these cells populate premandibular and pharyngeal regions, giving rise to skeletal elements like the trabeculae and Meckel's cartilage, which underpin the structural complexity absent in agnathans. The evolution of the peripheral nervous system (PNS) in vertebrates saw an expansion driven by neural crest-derived neurons and glia, supporting more complex sensory and autonomic functions essential for advanced behaviors. Trunk neural crest cells in early vertebrates contributed to sympathetic and enteric neurons, with diversification in gnathostomes allowing for elongated migration paths and integration into diverse circuits, such as those for and gut innervation. This expansion facilitated adaptations like enhanced sensory processing in aquatic and terrestrial environments. Neural crest adaptations have driven phenotypic across vertebrates, including pigment patterns in and amphibians, where multipotent crest cells generate melanophores, iridophores, and xanthophores for and signaling. In mammals, neural crest contributes to dental , with variations in arising from region-specific crest populations that pattern odontoblasts and enamel organs, enabling specialized feeding strategies. Comparatively, agnathans like the exhibit limited neural crest contributions, with restricted and fewer derivatives such as rudimentary ganglia, contrasting the extensive, multipotent crest in teleosts that supports and scale development. Genetic co-option underpinned these innovations, as neural crest gene regulatory networks (GRNs) were repurposed from ancestral border specifiers like Zic, /7, and genes, which were integrated into a vertebrate-specific module activating downstream effectors such as SoxE and . Recent evo-devo studies highlight neural crest involvement in limb , with crest-derived Schwann cells influencing fin patterning in teleosts through signaling crosstalk. In axolotls, neural crest contributions to regenerative niches, including peripheral glia, enhance limb regrowth capacity. Recent studies (2025) indicate that neural crest acquisition also facilitated the of the thyroid gland from the , enhancing endocrine complexity in vertebrates.

History

Discovery and early observations

The neural crest was first described in 1868 by Swiss embryologist Wilhelm His, who observed a band of cells, termed the "ganglion ridge" or Zwischenstrang, along the dorsal margins of the neural tube in chick embryos; he proposed these cells as the origin of spinal and cranial ganglia based on serial section reconstructions. In 1879, British anatomist Milnes Marshall coined the "neural crest" while studying olfactory organ development in vertebrates, describing it as an ectodermal ridge that delaminates and migrates to form peripheral ganglia, thereby emphasizing its role in early organization. Early 20th-century advances in experimental embryology further elucidated neural crest . In 1907, American zoologist Ross Granville Harrison pioneered techniques using frog neural tube explants, demonstrating active cellular outgrowth and patterns consistent with neural crest and movement away from the . By the 1920s, vital dye labeling experiments confirmed these origins and pathways; German embryologist Walter Vogt applied non-toxic dyes like Nile Blue to gastrulae and neurulae, mapping presumptive neural crest territories and tracing their contributions to ectomesenchyme and peripheral structures. Complementary work in chick embryos by researchers such as L.S. Stone used similar vital staining and transplantation to visualize routes and contributions to , establishing the neural crest as a distinct, migratory population arising transiently at the border. A key debate in the late 19th and early 20th centuries centered on the germ-layer origin of neural crest cells, with some researchers, influenced by classical germ-layer theory, attributing mesenchyme-like derivatives (e.g., craniofacial ) to rather than . This controversy, ignited by Julia Platt's 1893 observations of ectodermal contributions to mudpuppy skeletal elements, was resolved through heterotopic grafting experiments; studies by Lewis S. Stone (1926) and Carl P. Raven (1931) in amphibians showed that transplanted neural folds (ectodermal) generated donor-specific and pigment cells in host mesodermal environments, confirming the ectodermal origin and of neural crest cells. In the 1960s and 1970s, French developmental biologist Nicole Le Douarin advanced these findings using interspecific chimeras, where neural crest grafts into chick hosts were tracked via species-specific nuclear markers; these experiments definitively proved the multipotency of neural crest cells, as -derived cells populated diverse host derivatives including melanocytes, neurons, , and connective tissues across axial levels.

Key molecular and genetic advances

In the 1980s and early 1990s, key advances in understanding the epithelial-to-mesenchymal transition (EMT) essential for neural crest delamination came from the isolation of the Slug gene, a zinc finger transcription factor of the Snail family, which was shown to regulate cell behavior during vertebrate development, including neural crest emigration from the neural tube. This discovery established Slug as a critical driver of EMT, with antisense experiments demonstrating its necessity for neural crest migration in chick embryos. During the 1990s, genetic studies linked s in the to neural crest defects, notably through analysis of the Dominant (Dom) mouse model, where a in disrupted neural crest development and led to Hirschsprung disease-like phenotypes. Concurrently, human studies identified s in patients with Waardenburg-Hirschsprung syndrome (WS4), a neurocristopathy involving pigmentation, , and defects, confirming Sox10's role in neural crest specification and differentiation across species. The saw the development of (GRN) models that integrated multiple transcription factors and signaling pathways controlling neural crest formation, as proposed by Meulemans and Bronner-Fraser, who outlined hierarchical interactions among border specifiers like Tfap2 and FoxD3, and crest specifiers such as /10 and Snail2. These models highlighted the elucidation of and signaling pathways, where Wnt ligands promote neural crest induction at the border and BMP gradients regulate and migration, with combinatorial Wnt/BMP activity maintaining neural crest multipotency in and models. In the 2010s, CRISPR/Cas9 enabled precise knockouts of neural crest specifier genes, confirming their roles; for instance, targeted disruption of Tfap2a in chick embryos abolished border formation, while knockouts impaired and glial differentiation, validating the GRN hierarchy . Additionally, (iPSC)-derived neural crest stem cells (NCSCs) emerged as powerful tools for disease modeling, allowing generation of patient-specific NCSCs to recapitulate neurocristopathies like and through defects in migration and differentiation. Recent advances from the early 2020s onward have leveraged spatial transcriptomics to map neural crest migration dynamics, revealing spatiotemporal gene expression patterns during enteric neural crest cell wavefront progression in mouse embryos, including upregulated motility genes like Sema3a in leading cells. Epigenetic studies have further identified timing mechanisms in crest progenitors, with chromatin remodelers like Hmga1 showing bimodal roles in specification and delamination via CRISPR-validated knockouts, linking epigenetic barriers to developmental progression. Milestones include the 2016 recognition of EMT's broader developmental significance, building on Slug/Snail discoveries, and the advent of human organoid models integrating neural crest lineages to study diseases like DiGeorge syndrome, where ectodermal organoids reveal defective NC migration origins. Further, single-cell multi-omics and spatial transcriptomics have elucidated cranial neural crest patterning (2024), while studies on hominoid-specific transposable elements have shown their role in reshaping neural crest migration epigenomes (2025).

References

  1. [1]
    The Neural Crest - Developmental Biology - NCBI Bookshelf
    The cardiac neural crest cells can develop into melanocytes, neurons, cartilage, and connective tissue (of the third, fourth, and sixth pharyngeal arches). In ...
  2. [2]
    Neuroanatomy, Neural Crest - StatPearls - NCBI Bookshelf
    The neural crest is a collection of multipotent stem cells located at the side of the neural tube that migrate and give rise to many cell types.Introduction · Structure and Function · Embryology · Clinical SignificanceMissing: biology | Show results with:biology<|control11|><|separator|>
  3. [3]
    Evolution and Development of the Neural Crest: An Overview - NIH
    I. What is a neural crest cell? The neural crest is an embryonic cell type that is unique to vertebrates and forms numerous, diverse derivatives.
  4. [4]
    Neural crest stem cells: discovery, properties and potential for therapy
    Jan 10, 2012 · Neural crest (NC) cells are a migratory cell population synonymous with vertebrate evolution. They generate a wide variety of cell and tissue ...
  5. [5]
    The origin and evolution of vertebrate neural crest cells | Open Biology
    Jan 29, 2020 · The neural crest is a vertebrate-specific migratory stem cell population that generates a remarkably diverse set of cell types and structures.
  6. [6]
    Formation and migration of neural crest cells in the vertebrate embryo
    In the early embryo, the neural crest forms at the border between the neural plate, which will become central nervous system (CNS) and the non-neural ectoderm, ...
  7. [7]
    Specification and formation of the neural crest - PubMed Central - NIH
    The neural crest is an embryonic, multipotent cell population that migrates extensively and gives rise to a multitude of derivatives throughout the body, ...
  8. [8]
    Neural tube closure requires Dishevelled-dependent convergent ...
    Dec 15, 2002 · Convergent extension contributes directly to neural tube closure by narrowing the distance between the nascent neural folds.
  9. [9]
    The origins of the neural crest. Part I: embryonic induction
    Neural crest cells form at the border of the neural plate and epidermis, induced by local interactions between neural and non-neural ectoderm, possibly with ...
  10. [10]
    Pathways of trunk neural crest cell migration in the mouse embryo ...
    Apr 1, 1990 · ... timing of neural crest cell migration in mouse. In order to ... The first phase occurs between E8.5 and E9.5. Labelled cells leave ...
  11. [11]
    The use of human pluripotent stem cells for the in vitro derivation of ...
    During the development of the human embryo, transient structures such as the neural crest ... 3-4 weeks of gestation. Because of this early occurrence and ...
  12. [12]
    Comprehensive spatiotemporal analysis of early chick neural crest ...
    Transverse sections were performed on HH stage 8 (A'-J') and HH stage 10 (U'-DD') embryos as indicated. Gene expression in migrating neural crest cells is ...
  13. [13]
    The molecular basis of neural crest axial identity - PubMed Central
    The neural crest is a migratory cell population that contributes to multiple tissues and organs during vertebrate embryonic development.Cranial Neural Crest · Trunk Neural Crest · Sacral Neural Crest<|control11|><|separator|>
  14. [14]
    Relations and interactions between cranial mesoderm and neural ...
    Nov 2, 2005 · This review set out to highlight the progressive interactions between neural crest and mesoderm populations during craniofacial morphogenesis.
  15. [15]
    Current perspectives of the signaling pathways directing neural crest ...
    We provide a comprehensive review of the major signaling pathways that participate in neural crest induction, with a focus on recent developments and current ...
  16. [16]
    Neural crest multipotency and specification: power and limits of ...
    Apr 14, 2021 · ... Induction, specification and maturation of the territory that generates neural crest cells. Dev Biol. 2018; 444 Suppl 1: S36–S46. 10.1016/j ...
  17. [17]
    Gene bivalency at Polycomb domains regulates cranial neural crest ...
    Mar 31, 2017 · Epigenetic poising may allow the cranial NC cells to rapidly adapt their response to local variations in environmental signaling, potentially ...
  18. [18]
    Time to go: neural crest cell epithelial-to-mesenchymal transition
    Jul 29, 2022 · MMP9 likely modulates NCC EMT through its degradation of the adhesion protein CDH2 and membrane component laminin (Monsonego-Ornan et al., 2012) ...
  19. [19]
    The road best traveled: Neural crest migration upon the extracellular ...
    This breakdown in basement membrane is facilitated, in part, by neural crest cell-expressed proteases, which degrade the basement membrane to allow entry of ...Missing: paper | Show results with:paper
  20. [20]
    Neural crest delamination and migration: From epithelium-to ...
    Jun 1, 2012 · This review will summarize our current knowledge on delamination, EMT and migration of NC cells using key examples from chicken, Xenopus, zebrafish and mouse ...
  21. [21]
    Polarity and migration of cranial and cardiac neural crest cells
    The CXCR4/Sdf1 signaling pathway, originating from placodal cells, guides the migratory trajectories of NC cells. Meanwhile, C3a signaling, released by the NC ...
  22. [22]
    Control of neural crest cell behavior and migration - PubMed Central
    This study showed that trunk NCCs migrate in chainlike arrays through the rostral somite, with continuous filopodial contacts between them. Occasionally, ...
  23. [23]
    Neural Crest Migration Orchestrated by Molecular and Mechanical ...
    Oct 1, 2025 · Here, we review the guidance mechanisms involved in neural crest migration and stream formation. We first describe established concepts of ...
  24. [24]
    Cell extrusion drives neural crest cell delamination - PMC - NIH
    We identified, through live imaging of embryo development, that neural crest cells can also become migratory via cell extrusion, a mechanism thought to also be ...
  25. [25]
    In vivo confinement promotes collective migration of neural crest cells
    May 30, 2016 · We quantified these in streams along the anterior posterior axis of zebrafish and Xenopus embryos using time-lapse imaging and found that ...
  26. [26]
    Neurogenesis From Neural Crest Cells: Molecular Mechanisms in ...
    Cranial NCCs migrate to form sensory ganglia such as the trigeminal (V), the facial (VII), the glossopharyngeal (IX), the vagus (X) CN, and also to form the ...
  27. [27]
    Anatomy, Autonomic Nervous System - StatPearls - NCBI Bookshelf
    ... sympathetic ganglia. The parasympathetic innervation of the heart forms from the vagal neural crest.[42] The majority of the parasympathetic nervous system ...
  28. [28]
    Schwann cell precursors: where they come from and where they go
    Schwann cells have been suggested to derive from both multipotent and restricted neural crest progenitors. For example, some in vitro clones of quail ...
  29. [29]
    Schwann-cell differentiation in clonal cultures of the neural crest, as ...
    In particular, we found evidence for the existence of a bipotent precursor of Schwann cells and nonneuronal satellite cells; a common precursor of neurons, ...
  30. [30]
    The sacral neural crest contributes neurons and glia to the ... - PubMed
    The majority of the enteric nervous system is derived from vagal neural crest cells (NCC), which migrate to the developing gut, proliferate, form plexuses ...
  31. [31]
    Role of the neurotrophic factors BDNF and NGF in ... - PubMed - NIH
    The data suggest that BDNF, but not NGF, directs pluripotent neural crest cells to differentiate along the primary sensory neuron lineage.
  32. [32]
    Brain-derived neurotrophic factor stimulates survival and neuronal ...
    Brain-derived neurotrophic factor stimulates survival and neuronal differentiation in cultured avian neural crest. Brain Res. 1988 Jun 1;469(1-2):79-86. doi ...
  33. [33]
    Neural crest stem cells: discovery, properties and potential for therapy
    Jan 10, 2012 · The isolated postmigratory NCSC population differentiated into neurons, Schwann cells, and smooth muscle-like myofibroblasts in vitro; when ...Isolation Of Ncscs · Ncscs In The Skin · Figure 2<|separator|>
  34. [34]
    Neural Crest Stem Cells Persist in the Adult Gut but Undergo ...
    The persistence of NCSCs in the adult PNS opens up new possibilities for regeneration after injury or disease. ... stem cells also persist in the adult PNS.
  35. [35]
    The development, patterning and evolution of neural crest cell ...
    Neural crest cells then migrate extensively throughout the embryo, differentiating into a diverse array of cells, and contributing to a variety of tissues.Review Article · Patterning Of Neural Crest... · Neural Crest Cells And The...Missing: non- | Show results with:non-
  36. [36]
    Transcriptional and signaling regulation in neural crest stem cell ...
    Nov 11, 2008 · In this review, we discuss the known functional roles of these genes during NC stem cell-derived melanocyte development and propose alternative ...
  37. [37]
    Deep into melanocyte stem cells: the role of Kit signaling in their ...
    Jun 6, 2013 · The Kit receptor tyrosine kinase is a major extracellular signal pathway that controls many, if not all, cellular process of melanocytes.
  38. [38]
    signal transduction and transcription in the melanocyte lineage
    The aim of this review is to provide an overview of how gene expression in neural crest-derived melanocytes is controlled by the interplay between specific ...
  39. [39]
    The development of the chromaffin cell lineage from the neural crest
    Nov 17, 2009 · This review provides a summary of similarities and differences regarding the development of chromaffin cells and sympathetic neurons.Missing: origin | Show results with:origin
  40. [40]
    The development of the chromaffin cell lineage from the neural crest
    Nov 17, 2009 · The neuroendocrine chromaffin cells are located in the adrenal gland and in “extradrenal” paraganglia, as e.g. in the carotid body and “organ of ...
  41. [41]
    Cranial Neural Crest and Development of the Head Skeleton - NCBI
    For example, the ventral/distal element (V1) of the first arch (mandibular) forms Meckel's cartilage (mc), the template for the lower jaw, while the dorsal/ ...Introduction · Specification and Migration of... · Endoderm Patterns CNC and...
  42. [42]
    Periocular neural crest cell differentiation into corneal endothelium ...
    Jul 19, 2020 · During ocular development, periocular neural crest cells (pNC) migrate into the region between the lens and presumptive corneal epithelium ...Missing: connective | Show results with:connective
  43. [43]
    The cell-fate decision of dental follicle stem cells (DFSCs) with ...
    The dental follicle is a connective tissue derived from the neural crest and formed by ectomesenchymal progenitor cells during the cap stage. · In addition to a ...
  44. [44]
    Odontoblasts: the cells forming and maintaining dentine - PubMed
    Odontoblasts are tall columnar cells located at the periphery of the dental pulp. They derive from ectomesenchymal cells originated by migration of neural crest ...
  45. [45]
    Investigate the Odontogenic Differentiation and Dentin–Pulp Tissue ...
    Jun 5, 2020 · Our results showed that neural crest cells (O9-1 mouse cranial neural crest cell line) can sequentially differentiate into dentin matrix acidic ...
  46. [46]
    Smooth Muscle Cells Derived From Second Heart Field and Cardiac ...
    Jun 29, 2017 · Both cardiac neural crest– and second heart field–derived smooth muscle cells (SMCs) populate the media throughout the ascending aorta. •. In ...
  47. [47]
    SMAD4: A critical regulator of cardiac neural crest cell fate and ...
    Aug 31, 2023 · During embryogenesis, cardiac neural crest-derived cells (NCs) migrate into the pharyngeal arches and give rise to the vascular smooth muscle ...
  48. [48]
    Developmental dynamics of the neural crest–mesenchymal axis in ...
    May 13, 2022 · Neural crest cells differentiate into perivascular cells in the human prenatal thymus. To extend the analysis of the thymus stroma to human ...
  49. [49]
    Neural Crest‐Derived Mesenchymal Cells Support Thymic ...
    Nov 16, 2024 · Thymic mesenchymal cells originate from the Neural crest (NC) and mesoderm and contribute to thymus organogenesis, yet their role in thymic ...
  50. [50]
  51. [51]
    Neurocristopathy - an overview | ScienceDirect Topics
    Neurocristopathies refer to a spectrum of diseases associated with abnormal migration of neural crest cells, which can manifest in conditions such as ...Missing: pathophysiology | Show results with:pathophysiology
  52. [52]
    Systematic review of cardiovascular neurocristopathy ... - Frontiers
    Neurocristopathies are a group of genetic disorders that affect the development of cells derived from the NC. These could be from abnormalities in NCC migration ...
  53. [53]
    SOX10: 20 years of phenotypic plurality and current understanding ...
    The first clue that SOX10 is essential for development, especially in the neural crest, came with the discovery that heterozygous mutations occurring within and ...
  54. [54]
    SOX10 ablation severely impairs the generation of postmigratory ...
    More importantly, mutations in the human SOX10 gene are associated with several neurocristopathies, including demyelinating disorders and type 4 Waardenburg ...
  55. [55]
    Hirschsprung Disease - StatPearls - NCBI Bookshelf
    Jun 3, 2023 · ... prevalence varies from 1 to 1.63 per 10000 births ... Pathophysiology. Disturbed rostrocaudal migration of the neural crest ...
  56. [56]
    Neural crest development and disorders: from patient to model ...
    Jun 17, 2024 · Because of these properties, neurocristopathies (NCPs), which is the term used to classify genetic diseases associated with NC developmental ...
  57. [57]
    Emerging insights into cephalic neural crest disorders: A single ...
    Feb 3, 2024 · NEUROCRISTOPATHIES (NCPS) CLASSIFICATION​​ NCPs can be classified into dysgenetic form due to defective morphogenesis, neoplasms derived from ...Missing: pathophysiology | Show results with:pathophysiology
  58. [58]
    The origin and evolution of the neural crest - PMC - PubMed Central
    Thus, although some key tests remain, the evidence presented so far indicates that neural crest may well be an innovation of invertebrates, not of vertebrates.
  59. [59]
    Evolutionary crossroads in developmental biology: amphioxus
    Nov 15, 2011 · The neural crest cell GRN (NC-GRN) is partially conserved between amphioxus and vertebrates. The vertebrate NC-GRN includes 'border induction ...<|control11|><|separator|>
  60. [60]
    Insights from the amphioxus genome on the origin of vertebrate ...
    This discovery suggests that the evolutionary origin of neural crest in chordates may be more deeply rooted than previously thought. Given that amphioxus is ...
  61. [61]
    Migratory neuronal progenitors arise from the neural plate borders in ...
    Nov 19, 2015 · The neural crest is an evolutionary novelty that fostered the emergence of vertebrate anatomical innovations such as the cranium and jaws.
  62. [62]
    The Natural History of Model Organisms: Amphioxus as a ... - eLife
    Sep 18, 2023 · This new chordate phylogeny suggests an evolutionary explanation of why tunicates, despite their tremendous anatomical and genomic divergence, ...
  63. [63]
    Vertebrate innovations | PNAS
    First, head and trunk neural crest have different developmental fates and potentials, and it is not yet clear whether they evolved as a single cell population ...
  64. [64]
    Riding the crest to get a head: neural crest evolution in vertebrates
    May 9, 2023 · Here, we discuss recent findings regarding how neural crest cells may have evolved during the course of deuterostome evolution. The results ...
  65. [65]
    Evolution of the New Head by gradual acquisition of neural crest ...
    Apr 23, 2020 · The neural crest is a vertebrate innovation proposed to be a key component of the “New Head” that imbued vertebrates with predatory behavior. To ...
  66. [66]
    Ancient evolutionary origin of vertebrate enteric neurons from trunk ...
    Mar 20, 2017 · Our results suggest that neural crest-derived SCPs made an important contribution to the ancient ENS of early jawless vertebrates.
  67. [67]
    Neural crest cells as a source of microevolutionary variation - PMC
    Neural crest cells also produce pigment-producing cells. While mammals and birds only develop black/brown melanocytes, NCCs in fishes and amphibians generate a ...
  68. [68]
    Neural crest cells as a source of microevolutionary variation
    Neural crest cells (NCCs) are often touted as the “fourth germ layer” [1] ... defining characteristics like facial shape and coloration are generated ...
  69. [69]
    Significance of Heterochronic Differences in Neural Crest Cell ...
    The process of neural crest cell migration differs between the lamprey and swordtail [Hirata et al., Zoological Science 14: 305-312 (1997)].
  70. [70]
    Ancient Evolutionary Origin of the Neural Crest Gene Regulatory ...
    Sep 4, 2007 · These neural plate border specifiers in turn upregulate neural crest specifier genes, such as Slug/Snail, FoxD3, and members of the SoxE family.
  71. [71]
    A lamprey neural cell type atlas illuminates the origins of ... - Nature
    Sep 14, 2023 · Integrated comparative analyses of this atlas unveiled details of the cell type repertoire and molecular architecture of the ancestral ...
  72. [72]
    The Genetic Odyssey of Axolotl Regeneration: Insights and ...
    Dec 12, 2024 · The axolotl, a legendary creature with the potential to regenerate complex body parts, is positioned as a powerful model organism due to its ...
  73. [73]
    Control of Cell Behavior During Vertebrate Development by Slug, a ...
    Slug, a vertebrate gene encoding a zinc finger protein of the Snail family, is expressed in the neural crest and in mesodermal cells emigrating from the ...
  74. [74]
    Control of cell behavior during vertebrate development by Slug, a ...
    May 6, 1994 · Slug, a vertebrate gene encoding a zinc finger protein of the Snail family, is expressed in the neural crest and in mesodermal cells emigrating from the ...
  75. [75]
    SOX10 mutation disrupts neural crest development in Dom ... - Nature
    Jan 1, 1998 · SOX10 mutation disrupts neural crest development in Dom Hirschsprung mouse model ... Another mouse model of HSCR disease, Dom, arose ...
  76. [76]
    SOX10 mutations in patients with Waardenburg-Hirschsprung disease
    Feb 1, 1998 · Here we show that patients from four families with WS4 have mutations in SOx10, whereas no mutation could be detected in patients with HSCR alone.<|control11|><|separator|>
  77. [77]
    Neural crest stem cell maintenance by combinatorial Wnt and BMP ...
    Our study provides multiple lines of evidence for a cross-talk between Wnt and BMP signaling in eNCSCs. Wnt1 treatment as well as sustained β-catenin activity ...Missing: elucidation | Show results with:elucidation
  78. [78]
    Optimization of CRISPR-Cas9 genome editing for loss-of-function in ...
    We test the ability of our optimized CRISPR/Cas9 system to knock out key transcription factors in the neural crest, including the neural plate border specifier ...
  79. [79]
    Human Pluripotent Stem Cell-Derived Neural Crest Cells for Tissue ...
    Feb 22, 2019 · Neural crest cells (NCCs) are a multipotent and migratory cell population in the developing embryo that contribute to the formation of a ...
  80. [80]
    Bimodal function of chromatin remodeler Hmga1 in neural crest ...
    Sep 23, 2020 · Our results show that Hmga1 functions in a bimodal manner during neural crest development to regulate specification at the neural plate border, and subsequent ...<|control11|><|separator|>
  81. [81]
    Human ectodermal organoids reveal the cellular origin of DiGeorge ...
    Aug 8, 2025 · Neurocristopathies account for half of all birth defects and several cancers highlighting the need to understand early neural crest (NC) ...