Fact-checked by Grok 2 weeks ago

Gastrulation

Gastrulation is a pivotal in early embryonic development wherein the blastula, a single-layered hollow sphere of undifferentiated cells, undergoes coordinated morphogenetic movements to reorganize into the gastrula, a multilayered structure defined by the formation of three primary germ layers: , , and . These germ layers represent the foundational cellular populations that differentiate into all subsequent tissues and organs, with giving rise to the and , to muscles, bones, and circulatory elements, and to the linings of the digestive and respiratory tracts. The process is characterized by dynamic cellular behaviors including , where cells fold inward to form the ; , the migration of cells over the lip of the blastopore; and ingression, the individual movement of cells into the interior—mechanisms that collectively establish bilateral symmetry and body axes while varying across taxa such as in the distinct mouth-first () versus anus-first () patterns observed in and vertebrates, respectively. In mammalian embryos, gastrulation initiates around the third week post-fertilization, driven by signaling gradients like those involving , Wnt, and Nodal pathways that induce epithelial-to-mesenchymal transitions essential for and specification, underscoring its conserved role in metazoan evolution despite mechanistic diversity. Disruptions in these precisely timed events can lead to congenital malformations, highlighting gastrulation's critical causality in developmental fidelity.

Fundamentals

Definition and Core Process

Gastrulation is the phase of early embryonic development in which the blastula—a hollow sphere consisting of a single layer of cells enclosing a blastocoel—is transformed into the gastrula, a multilayered structure featuring three primary germ layers: ectoderm, mesoderm, and endoderm. This reorganization occurs through coordinated cellular behaviors that internalize specific cell populations, establishing the basic body plan of the embryo. The core process involves distinct morphogenetic movements, such as invagination (inward folding of the cell sheet), ingression (individual cells delaminating and migrating inward), epiboly (expansion and thinning of the outer layer), involution (cells rolling inward at the margin), and convergence-extension (tissue narrowing and elongation). These actions, often initiated at a specific site like the vegetal pole in invertebrates or the primitive streak in vertebrates, rely on epithelial-to-mesenchymal transitions to enable cells to ingress and displace into interior positions, forming the inner endoderm and middle mesoderm beneath the protective ectoderm. Germ layer formation during gastrulation positions ectoderm as the outermost layer destined for epidermal and neural tissues, mesoderm as the intermediate layer for connective, muscular, and vascular structures, and endoderm as the innermost layer for digestive and respiratory epithelia. This trilaminar organization, conserved across metazoans despite mechanistic variations, also orients embryonic axes (e.g., dorsal-ventral, anterior-posterior) critical for further differentiation.

Etymology

The term gastrulation was coined by German biologist in 1872, in his work Die Kalkschwämme (Biology of Calcareous Sponges), to describe the embryonic process leading to the formation of the primitive gut. It derives from the gastḗr (γαστήρ), meaning "stomach" or "belly," reflecting the and reorganization that establish the , the precursor to the digestive tract. The noun gastrula, denoting the resulting embryonic stage, combines this root with the diminutive suffix -ula.

Biological and Evolutionary Importance

Gastrulation establishes the foundational three primary germ layers—ectoderm, mesoderm, and endoderm—which differentiate into all major tissue types and organs in triploblastic animals, thereby initiating the of the . This process reorganizes the blastula's uniform cell layer into a structured gastrula, specifying and positioning these layers to enable subsequent and . By generating spatial organization, including anterior-posterior and dorsal-ventral axes, gastrulation ensures directional development critical for embryonic viability. Biologically, gastrulation's cell rearrangements, such as and convergence-extension, are essential for transitioning from a simple spherical to a complex form capable of supporting differentiated functions, with disruptions often resulting in lethal developmental arrest or congenital anomalies. The formation of the , in particular, allows for the development of supportive structures like muscles and circulatory systems, underscoring gastrulation's role in enabling multicellular complexity beyond mere . Evolutionarily, gastrulation exhibits deep conservation across metazoans, with core morphogenetic movements and regulatory networks—such as those involving , Wnt, and Nodal signaling—predating the protostome-deuterostome divergence, indicative of shared ancestry in bilaterian lineages. While modes vary (e.g., spiraling in protostomes versus radial in deuterostomes), the underlying genetic toolkit for specification remains homologous, suggesting that evolutionary primarily modified deployment rather than inventing new mechanisms . This conservation highlights gastrulation as a pivotal in metazoan evolution, facilitating the scalable complexity of animal body plans from simple to vertebrates.

Morphogenetic Mechanisms

Fundamental Cell Movements

Gastrulation relies on a set of fundamental movements that reorganize the blastula into a multilayered gastrula, establishing the , , and germ layers through coordinated , spreading, and reshaping of cell sheets. These movements, conserved across metazoans but varying in execution, include , , ingression, , , and convergence-extension, often involving changes in cell shape, , and driven by cytoskeletal dynamics and interactions. In vertebrates, these processes typically initiate at a specified , such as the blastopore lip in amphibians, where cells undergo epithelial rearrangements to internalize presumptive mesendoderm. Invagination entails the inward folding of an epithelial cell sheet, akin to dimpling a flexible , which creates a pouch-like structure that contributes to cavity formation or tissue internalization. This movement is prominent in embryos, where primary cells invaginate at the vegetal pole to form the , and in gastrulation at the dorsal blastopore lip. It relies on apical constriction of cells via actomyosin contractility, reducing surface area and driving buckling without requiring widespread . Involution involves the inward rolling of a coherent epithelial layer over an edge, such as cells from the marginal zone folding under the roof in embryos to displace presumptive and internally. This movement, distinct from by its sheet-like migration around a or rim, extends the involuting layer to line embryonic cavities and is regulated by planar cell polarity and Wnt signaling. Involution facilitates the transition from superficial to deep layers, ensuring proper positioning. Ingression occurs when individual epithelial cells detach from a sheet, undergo , and migrate independently into the interior, often as that later reintegrate into tissues. This is exemplified by primary mesenchyme ingression in echinoderms, where cells bottle-shape and delaminate via downregulation of E-cadherin and upregulation of , contributing to . Ingression allows dispersed cell populations to populate spaces between germ layers, contrasting with collective sheet movements. Delamination separates cells from an epithelial layer into basal or apical sublayers, or as individuals, without extensive , often preceding other movements to refine stratification. Seen in avian primitive streak formation, it involves partial and is crucial for generating multilayered epithelia from monolayers. Epiboly describes the thinning and spreading of an external epithelial sheet to envelop the , driven by radial intercalation and membrane expansion, as in where blastoderm cells expand over the cell. This movement increases surface area coverage, coordinating with internalization to enclose or internalize cells. Convergence-extension reshapes tissues by cells converging toward the midline while intercalating and elongating perpendicularly, narrowing and lengthening the along the anteroposterior . Powered by polarized protrusions and non-canonical Wnt/ signaling, it is essential in formation and amplifies axial structures during gastrulation. These movements collectively ensure efficient segregation and embryonic establishment, with variations reflecting evolutionary adaptations.

Germ Layer Formation and Gastrula Structure

Gastrulation establishes the three primary germ layers—ectoderm, mesoderm, and endoderm—through coordinated cellular rearrangements that convert the blastula's single epithelial layer into a trilaminar structure. The comprises the presumptive outer layer, destined to form epidermal tissues and the ; the arises as an intermediate layer generating connective tissues, muscles, and circulatory elements; and the internalizes to line the primitive digestive cavity, giving rise to gut and associated organs. These layers emerge via processes such as ingression and , where cells from the surface layer migrate inward, displacing or supplementing prior hypoblast-like cells in vertebrates. A key mechanism in germ layer formation is the epithelial-to-mesenchymal transition (EMT), during which epithelial cells of the epiblast lose cell-cell , apical-basal , and acquire migratory mesenchymal properties, enabling their contribution to and . This transition, regulated by signaling pathways like and Wnt, allows cells to ingress through structures such as the in amniotes or the blastopore in other taxa, positioning mesodermal precursors laterally and endodermal cells medially. Post-EMT, some cells undergo mesenchymal-to-epithelial transition to reform epithelial sheets, stabilizing layer identities. The resulting gastrula exhibits a characteristic structure with the enveloping the externally, the interspersed as loosely organized sheets or , and the forming a continuous internal lining around the —a fluid-filled connected to the exterior via the blastopore. This blastopore marks the site of cell internalization and defines embryonic axes, with the serving as the precursor to the gut. In triploblastic animals, the trilaminar organization establishes the foundational , with germ layer positions reflecting conserved morphogenetic principles across metazoans.

Molecular Regulation

Key Signaling Pathways

The molecular orchestration of gastrulation relies on conserved signaling pathways that integrate environmental cues, cell-cell interactions, and transcriptional responses to drive specification and morphogenesis. Principal pathways include members of the transforming growth factor-β (TGF-β) superfamily such as Nodal, which initiates formation; Wnt/β-catenin signaling, which promotes posterior identity; (BMP) signaling, which establishes ventral fates; and (FGF) signaling, which supports and survival during . These pathways exhibit spatiotemporal dynamics, with mutual antagonism and synergy ensuring precise patterning, as disruptions in any one can arrest gastrulation progression. Nodal signaling, part of the TGF-β family, is pivotal for inducing the primitive streak and specifying mesendodermal progenitors in vertebrates. In mice and amphibians, Nodal ligands secreted from extraembryonic or vegetal regions activate Smad2/3 transcription factors via receptors like Alk4/5/7, promoting brachyury (T) expression and epiblast for gastrulation entry as early as embryonic day 6.5 in mice. Antagonists such as Lefty and refine Nodal gradients to prevent ectopic induction, with genetic knockouts demonstrating that Nodal-null embryos fail to form by E7.5. Wnt/β-catenin signaling cooperates with Nodal to posteriorize the embryo and stabilize mesodermal fates during streak formation. Canonical Wnt ligands (e.g., Wnt3 in mice) inhibit GSK3β, leading to β-catenin nuclear accumulation and activation of targets like Tbx6 and Fgf8, which are essential for paraxial development; in , maternal Wnt8 reinforces at shield stage (5.25 hours post-fertilization). Non-canonical Wnt pathways, such as Wnt/PCP, further regulate convergent extension movements by polarizing cytoskeletal dynamics via RhoA and JNK. BMP signaling, also TGF-β-related, patterns the dorsoventral axis by promoting ventral and lateral while being antagonized dorsally by secreted inhibitors like Chordin, Noggin, and from the organizer region. In , BMP4 gradients peak ventrally at stage 10.5, driving ventrolateral fate via Smad1/5/8; loss-of-function studies show uniform dorsalization in BMP receptor mutants. Interactions with Wnt and Nodal modulate BMP thresholds, ensuring compartmentalized fates during gastrulation. FGF signaling sustains mesodermal gene expression and facilitates epithelial-mesenchymal transition (EMT) and migration post-involution. FGF8 and FGF4 from the primitive streak activate ERK/MAPK cascades, inducing snail and twist for EMT while preventing apoptosis; in chick embryos, FGF inhibition halts head process extension by HH stage 4. Crosstalk with BMP inhibits neural induction, reinforcing ectodermal competence boundaries. These pathways are evolutionarily conserved yet modulated by context-specific regulators, with quantitative models revealing threshold-dependent responses that underpin robust gastrulation across taxa.

Genetic and Epigenetic Controls

Genetic of gastrulation is mediated by hierarchical gene regulatory networks (GRNs) that coordinate activity with extracellular signaling to specify germ layers and drive morphogenetic movements. These networks integrate inputs from pathways like Wnt, Nodal, and to activate lineage-specific , with conserved such as Brachyury (T-box family) essential for and axial in vertebrates. GATA4/5/6 factors similarly function across species to regulate and formation, binding enhancers that respond to nodal signaling for timely activation during cell ingression. Disruptions in these GRNs, as seen in Trim71 mutants, lead to dysregulated expression of mesodermal , impairing gastrulation progression and highlighting the precision of transcriptional hierarchies. A core set of approximately 75 transcription factors forms a conserved regulatory module across mammalian gastrulation, controlling epiblast and formation through shared cis-regulatory syntax. This module, identified via single-cell atlases, underscores evolutionary stability despite species-specific timing, with factors like Sox17 and Foxa2 enforcing endodermal identity post-ingression. In protostomes and deuterostomes, analogous TFs such as and in orchestrate mesoderm invagination, reflecting modular GRN conservation amid divergent morphologies. Epigenetic controls complement genetic programs by dynamically remodeling to permit or restrict access to developmental loci during the pluripotent-to-multipotent transition. patterns shift globally in mouse epiblasts prior to gastrulation, with demethylation at bivalent promoters enabling poised expression of lineage genes like those in the T/Brachyury network. modifications, including enrichment by Polycomb repressive complexes at CpG islands, maintain repression of non-gastrulation fates, while Trithorax-mediated activates mesendodermal enhancers as cells ingress. These marks interact cooperatively; for instance, low correlates with active , facilitating rapid transcriptional responses to signaling gradients. Chromatin accessibility assays reveal gastrulation-stage surges in open regions near TFs like Gata6, correlating with repositioning and variant incorporation to stabilize identities. In human models mimicking gastrulation, epigenetic profiling shows conserved principles of / bivalency resolution, directing specification alongside somatic lineages. Such ensures irreversible commitment, as evidenced by elevated epigenetic age minima at gastrulation onset, underscoring its role as a causal bottleneck for developmental fidelity.

Comparative Gastrulation Across Taxa

Protostomes Versus Deuterostomes

A primary distinction between s and s arises during gastrulation with the fate of the blastopore, the initial opening formed in the embryo. In protostomes, the blastopore develops into the , reflecting a "first mouth" developmental pattern. In contrast, deuterostomes exhibit the blastopore becoming the anus, with the forming secondarily from a separate opening. This difference, observed consistently across major taxa such as annelids and mollusks for protostomes versus echinoderms and chordates for deuterostomes, underscores divergent strategies in gut formation and anterior-posterior axis establishment./13%3A_Module_10-_Animal_Diversity/13.21%3A_Embryological_Development) Preceding gastrulation, cleavage patterns further differentiate the groups, influencing subsequent morphogenetic movements. Protostomes typically undergo spiral cleavage, where daughter cells divide at oblique angles, resulting in a skewed arrangement relative to underlying cells and promoting determinate with early cell fate commitment./13%3A_Module_10-_Animal_Diversity/13.21%3A_Embryological_Development) Deuterostomes, however, feature radial cleavage, with divisions parallel or perpendicular to the polar , aligning cells in tiers and supporting indeterminate where early blastomeres retain regulative potential./13%3A_Module_10-_Animal_Diversity/13.21%3A_Embryological_Development) These patterns set the stage for gastrulation, where invagination often emphasizes mesendoderm migration toward the blastopore to form the mouth-first , while processes prioritize radial symmetry in reorganization. Coelom formation, which emerges post-gastrulation from mesodermal precursors, also varies mechanistically between the clades. Protostomes employ , wherein the arises by splitting solid mesodermal masses into cavities. Deuterostomes utilize enterocoely, involving outgrowths or pouches from the wall that pinch off to form coelomic spaces. These modes correlate with the blastopore's role: schizocoely aligns with originating laterally to the blastopore-derived mouth, whereas enterocoely ties to mesodermal evagination near the blastopore-anus site. Such differences impact flexibility, with regulative mechanisms allowing greater evolutionary adaptability in organ positioning compared to the more fixed mosaic development./13%3A_Module_10-_Animal_Diversity/13.21%3A_Embryological_Development) While these traits define the protostome-deuterostome dichotomy, exceptions exist, such as variable blastopore fates in some protostomes, challenging strict delineations and prompting ongoing phylogenetic reevaluations based on molecular data./13%3A_Module_10-_Animal_Diversity/13.21%3A_Embryological_Development) Nonetheless, gastrulation's morphological signatures remain key for classifying bilaterian animals, informing evolutionary divergence estimated around 550-600 million years ago during the Ediacaran-Cambrian transition.

Invertebrate Models: Sea Urchins

Sea urchins, particularly species like Strongylocentrotus purpuratus, serve as a premier model for studying gastrulation due to their transparent embryos, external , and to produce large numbers of synchronously cleaving eggs that are easily fertilized . These features have facilitated over 150 years of research, enabling detailed observation of cellular and molecular events without invasive techniques. As s, sea urchins exhibit gastrulation patterns homologous to vertebrates, including radial cleavage and formation of the from the blastopore, contrasting with mouth-first . Gastrulation commences at the blastula stage, around 9-10 hours post-fertilization at 15°C, with the vegetal plate cells initiating primary mesenchyme cell (PMC) ingression. PMCs, derived from micromeres at the fourth , undergo epithelial-to-mesenchymal transition (), detach from the vegetal via apical constriction and loss of cell-cell adhesions, and ingress into the using for . These cells, numbering about 20-40 per , settle at the animal pole and vegetal ridge to secrete the larval , guided by chemotactic signals such as VEGF and FGF. Disruption of these pathways, as shown in experiments, arrests PMC and skeletal formation. Concurrent with or following PMC ingression, the remaining vegetal plate undergoes primary to form the , the precursor to the gut. This involves convergent extension of the endodermal , where bottle cells at the vegetal margin constrict apically, driving a purse-string-like folding, while contractility and remodeling facilitate tissue bending without requiring oriented . The then elongates through secondary mesenchyme cell (SMC) addition and filopodial traction, propelled by polarized actomyosin flows and adhesion to the roof. By late gastrulation, around 24-30 hours, the tip contacts the oral , establishing the mouth via fusion, while SMCs contribute to coelomic pouches and pigment cells. Gene regulatory networks (GRNs) orchestrate these movements, with transcription factors like ets1, alx1, and tbr specifying PMC fate and , activated downstream of Delta-Notch signaling from micromeres. relies on goosecoid and brachyury for epithelial integrity and elongation, modulated by Nodal, , and Wnt pathways that pattern the vegetal-oral axis. These networks, mapped through perturbation and cis-regulatory , reveal robust linkages between and , as PMCs ingress even in isolated cells under specific conditions. studies have thus illuminated conserved mechanisms, such as and convergent extension, applicable to gastrulation while highlighting echinoderm-specific adaptations like skeletal .

Vertebrate Models: Amphibians

In amphibians such as Xenopus laevis, gastrulation transforms the spherical blastula into a multilayered gastrula through precise cell rearrangements that establish the , , and germ layers, with the process initiating at stage 10 (approximately 8-10 hours post-fertilization at 23°C). The dorsal blastopore lip emerges first as a thickened region in the marginal zone, where bottle cells—narrow, flask-shaped cells derived from superficial and deep marginal layers—undergo epithelial-to-mesenchymal transition () and ingress to initiate , forming the blastopore rim that serves as the portal for mesendodermal internalization. Presumptive from the vegetal hemisphere and from the equatorial marginal zone involute through this lip, displacing the cavity and expanding the , a primitive gut cavity lined by . Coordinated morphogenetic movements drive layer formation: thins and spreads the prospective ectodermal animal cap vegetally over the , covering up to 60-70% of the surface by mid-gastrulation; directs deep marginal cells inward along the roof via directed migration and traction forces; and convergence-extension intercalates mediolateral cells to elongate the anteroposterior axis by 2-3 fold while narrowing the mediolateral dimension, generating tensile forces that peak at 1.5-2.0 μN during early gastrulation and exceed 4.0 μN by onset. These rearrangements position between and , with dorsal fating to and somites, ventral to blood and lateral plate, and lateral to pronephros, as traced by vital dye labeling experiments showing 80-90% fidelity in cell . The Spemann-Mangold organizer, identified in newt embryos in 1924 and homologous in Xenopus at the dorsal blastopore lip, functions as a signaling center by secreting BMP antagonists like chordin and noggin, which dorsalize ventral mesoderm and induce neural ectoderm in overlying tissues, as demonstrated by transplantation assays inducing secondary axes with 70-100% efficiency depending on graft timing. Non-canonical Wnt/PCP signaling, involving RhoA GTPases and myosin II contractility, mediates convergence-extension independently of canonical Wnt/β-catenin pathways, with disruptions via dominant-negative constructs reducing extension by 50% or more in explants. Cell adhesion molecules such as cadherins and integrins facilitate differential migration, with calcium-dependent adhesion gradients ensuring tissue integrity during shear stresses up to 10-20 dyn/cm². EMT underlies bottle cell formation and involuting mesoderm motility in amphibians, enabling mesenchymal migration before reversion to epithelial states in target tissues.

Vertebrate Models: Amniotes and Mammals

In amniote embryos, gastrulation proceeds through the formation of a primitive streak, a transient structure that serves as the site for epithelial-to-mesenchymal transition (EMT) and ingression of epiblast cells to generate mesoderm and definitive endoderm. This process establishes bilateral symmetry and the anteroposterior axis, with conserved roles for Wnt, Nodal, BMP, and FGF signaling pathways across amniotes. The chick embryo provides a primary model for yolk-rich amniotes, where the epiblast and hypoblast form prior to streak initiation around 15 hours post-laying. In chicks, formation begins posteriorly near Koller's sickle, a region of cell intercalation that induces streak elongation anteriorly through epithelial rearrangements and proliferation, reaching full extension by Hamburger-Hamilton stage 4 (approximately 18-19 hours of incubation). , an extraembryonic layer, positions the streak by secreting antagonists such as , which inhibit Nodal and signaling to prevent ectopic streak formation.00318-0) Ingression occurs via at the streak, with cells migrating laterally and anteriorly to displace hypoblast-derived , forming paraxial , lateral plate, and axial structures like Hensen's node at the anterior terminus.00041-1) Cell movements include midline and counterrotational flows, driven by polarized intercalations. Mammalian gastrulation, exemplified by the , shares the mechanism but adapts to a yolk-poor , initiating around embryonic day 6.25 (E6.25) at the distal-posterior epiblast without a equivalent. The anterior visceral (AVE) migrates posteriorly to break symmetry and restrict streak formation, guided by Nodal inhibition via Lefty and Cer1. Streak extension to E7.5 involves Wnt3-mediated and proliferation, with ingressing cells forming mesenchymal that spreads as wings between epiblast and visceral . Definitive emerges via subsequent mesenchymal-to-epithelial transition (MET) and intercalation into extraembryonic by E8.75, while diversifies into axial, paraxial, intermediate, and lateral subtypes based on expression and signaling gradients. Unlike , mouse ingression lacks pronounced convergent extension, relying more on local EMT without large-scale posterior convergence. Key differences between and mammalian models include dependency—chick gastrulation occurs on a yolky disc with involvement, whereas mouse proceeds in a cup-shaped structure with AVE-directed patterning—and dynamic flows, with chicks exhibiting stronger rotational movements absent in mice. Both, however, demonstrate regulative , as evidenced by streak after . In humans, appearance around day 14 post-fertilization mirrors mouse timing proportionally, though direct observation is limited.

Evolutionary Perspectives and Debates

Historical Research Milestones

In 1817, Christian Heinrich Pander published observations on chick embryo development, identifying the formation of three distinct germ layers—ectoderm, mesoderm, and endoderm—from the blastoderm, providing the initial empirical basis for recognizing gastrulation as the morphogenetic process that establishes these foundational tissues. This trilaminar organization was later generalized by Karl Ernst von Baer, who in 1828 extended the germ layer concept to all vertebrates through comparative studies, emphasizing conserved developmental patterns across species and linking them to organ formation during gastrulation. Ernst Haeckel coined the terms "gastrula" and "gastrulation" in 1872 while studying development, describing the process as an forming a primitive gut-like structure analogous across metazoans, and in 1874 proposed the gastraea hypothesis positing a hypothetical with gastrula-like organization to explain evolutionary conservation of origins. These conceptual advances shifted focus from descriptive to dynamic cellular rearrangements, though Haeckel's recapitulationist interpretations later faced scrutiny for overemphasizing phylogenetic in . Experimental embryology advanced in the early with vital techniques; Walter Vogt applied neutral red and Nile blue dyes to blastulae in 1923–1929, producing the first comprehensive fate maps that traced presumptive territories and quantified movements during gastrulation, revealing mesoderm's dual superficial and deep origins. Concurrently, in 1924, and Hilde Mangold demonstrated that the blastopore lip in gastrulae acts as an organizer, inducing secondary axes and neural via host-graft transplants, a discovery earning Spemann the 1935 and establishing as a causal mechanism coordinating specification and . Mid-20th-century refinements included Johannes Holtfreter's 1943–1944 exogastrulation experiments on embryos, which dissociated induction from morphogenetic movements, showing that presumptive and could self-differentiate while required signals, thus clarifying gastrulation's regulative versus mosaic elements. These milestones, grounded in direct observation and manipulation, resolved early debates on preformation versus epigenesis, privileging causal interactions over rigid homologies.

Debates on Germ Layer Homology and Origins

The evolutionary origins of and , the inner s formed during gastrulation, have been debated for decades, with uncertainty surrounding their ancestral specification and interrelationship in early metazoans. Comparative analyses of transcription factors such as GATA4-6, , , and Brachyury indicate that these genes, which regulate formation in bilaterians, likely evolved from roles in basic cellular processes like and , supporting a gradual emergence rather than abrupt invention. In diploblastic cnidarians, the inner cell layer expresses homologs of bilaterian endomesodermal markers, prompting over whether this reflects to endoderm or convergent co-option of ancient regulatory networks. Sponge development provides limited support for germ layer homology, as embryonic cell layers in species like Amphimedon queenslandica exhibit transient patterning without fixed identities akin to eumetazoan , , or ; instead, cells transdifferentiate flexibly post-embryogenesis. GATA expression in larval inner layers hints at a shared endomesodermal precursor, but overall evidence favors gastrulation as an eumetazoan innovation after the poriferan divergence around 600-800 million years ago. Within triploblastic bilaterians, homology is inferred from conserved topological arrangements and regulatory networks, despite divergent cellular origins—such as endomesodermal ingression in many protostomes versus inductive separation in deuterostomes. Shifts in germ layer contributions, exemplified by variable versus mesodermal origins of vertebrate skull sutures (e.g., as neural crest-mesoderm in mice but mesoderm-mesoderm in ), necessitate homology criteria beyond origin, including inductive signaling centers and evolutionary continuity. Emerging molecular data challenge rigid germ layer boundaries, as ectodermal gonopores in xenacoelomorphs (basal bilaterians) deploy endodermal hindgut markers like caudal, brachyury, and Wnt signaling, suggesting functional to bilaterian anuses despite disparate origins and implying that strict endoderm-ectoderm distinctions may obscure deeper developmental equivalences. Such findings underscore ongoing debates on whether triploblasty arose via endodermal splitting or mesoderm insertion, with implications for reconstructing the .

Challenges from Non-Model Organisms

Non-model organisms, encompassing taxa beyond standard laboratory species such as , Xenopus laevis, and Mus musculus, exhibit gastrulation processes that diverge markedly in morphology, cell movements, and molecular regulation, complicating extrapolations from model systems. For instance, in cnidarians like Nematostella vectensis, embryos typically undergo for internalization, but experimental cell dissociation induces a switch to multipolar ingression or , demonstrating context-dependent plasticity not routinely observed in bilaterian models. Similarly, arthropods such as Chironomus riparius favor ingression over the seen in , with maternal factors like /t48 mRNA dictating the mode, highlighting how subtle genetic variations yield alternative outcomes. In vertebrates, non-model species further underscore this variability; catsharks (Scyliorhinus spp.) form flat gastrulation discs with crescent-shaped ingressions adapted to large yolk reserves, differing from the ring-like and in amphibians or ingression in teleosts. reptiles employ bi-modal strategies, combining streak-like ingression with yolk-constrained adaptations, challenging direct to or mammalian streaks. Among mammals, rabbits and humans deviate from mice in gastrulation tempo and signaling: human requires SNAI2 expression and MEK-dependent epithelial-mesenchymal transition (), absent in , while somitogenesis periodicity differs (e.g., ~3 hours in humans versus ~2 hours in mice) due to variations in HES7 protein stability. These observations pose technical challenges, including opaque embryos hindering live imaging, protracted developmental timelines, and paucity of genetic tools like transgenics, which limit functional validation of mechanisms in species such as lampreys or bichirs. Conceptually, non-bilaterian non-models like ctenophores and cnidarians lack a distinct layer, with and often arising via or polyclism rather than canonical gastrulation ingressions, fueling debates on and the evolutionary origins of triploblasty. Such diversity implies that yolk geometry, signaling thresholds (e.g., FGF/ gradients), and intrinsic biochemical rates drive adaptive innovations, rather than rigid , necessitating broader comparative datasets to refine causal models of formation.

In Vitro and Synthetic Models

Development of Gastruloids and Engineered Systems

Gastruloids are three-dimensional aggregates of pluripotent stem cells, such as embryonic stem cells (mESCs) or embryonic stem cells (hESCs), engineered to self-organize and recapitulate key aspects of post-implantation mammalian gastrulation, including , formation, and specification. The first 3D gastruloids were generated in by aggregating approximately 300 mESCs in low-adherence conditions, yielding structures that elongated along an anterior-posterior axis within 5-7 days under serum-free media supplemented with N2B27 and minimal growth factors to promote without exogenous morphogens. Concurrently, two-dimensional engineered models emerged using micropatterned substrates to confine hESCs to circular adhesive islands (0.5-1 mm diameter) coated with , enabling patterned driven by uniform BMP4 exposure, which induced brachyury-expressing primitive streak-like cells at colony edges and subsequent patterning. Protocols for 3D gastruloid generation typically involve dissociating pluripotent cells into single-cell suspensions via or Accutase, followed by plating at densities of 200-500 cells per in U-bottom 96-well low-attachment plates to form embryoid body-like structures. Culture proceeds in defined media activating endogenous signaling pathways—such as WNT, , NODAL, and FGF—often starting with Activin A and WNT agonists for induction, transitioning to BMP4 for mesendoderm specification, with aggregates reaching 500-1000 μm in size by day 4-5 and exhibiting formation and axial elongation by day 7. For human models, similar aggregation uses hESCs or induced pluripotent cells (iPSCs) under feeder-free conditions, with ethical guidelines limiting progression beyond 14 days or implantation potential. Engineered enhancements refine these systems for spatiotemporal control, incorporating micropatterned (PDMS) substrates or bioengineered microwell arrays to impose geometric constraints that influence and tissue . Microfluidic devices deliver precise gradients, mimicking signaling asymmetries, while synthetic biology tools—such as CRISPR-edited lines expressing inducible transgenes for pathway modulation—enable dissection of causal mechanisms like epithelial-mesenchymal transition during ingression. Chimeric gastruloids, formed by mixing differentially labeled populations, further allow lineage tracing and perturbation studies, revealing non-cell-autonomous interactions in allocation. These advancements, building on initial self-organizing aggregates, have scaled production to thousands of uniform structures for , though models remain limited by absent extraembryonic support and incomplete somitogenesis.

Achievements and Empirical Limitations

Gastruloids, aggregates of pluripotent cells that self-organize to mimic early gastrulation, have successfully recapitulated key aspects of specification and patterning in mammalian models. For instance, gastruloids derived from embryonic cells exhibit primitive streak-like structures, epiblast , and differentiation into the three layers within 72-96 hours post-aggregation, enabling of signaling pathways such as Wnt and Nodal. Recent protocols incorporating pulses have induced posterior axial elongation and somitogenesis-like oscillations, producing trunk-like structures with segmented by day 5-7, thus modeling post-gastrulation . These systems facilitate genetic perturbations, revealing causal roles for metabolites like in induction, as demonstrated in large-scale metabolomic analyses of gastruloids. Engineered variants, including vascularized gastruloids, have further advanced modeling of interactions, such as cardiac specification and primitive vascular networks emerging by day 4-6 in systems. Microraft technologies have scaled gastruloid to thousands per experiment, allowing image-based phenotyping and for aberrant morphologies, which supports quantitative studies of developmental robustness and genetic variants. Such platforms have quantified spatiotemporal , confirming driven by diffusible morphogens without external scaffolds, thereby validating intrinsic cellular programs . Despite these advances, gastruloids exhibit empirical limitations in faithfully replicating gastrulation dynamics. Basic models lack extraembryonic tissues, resulting in incomplete anterior-posterior axis formation and absent structures like the anterior visceral endoderm, which restricts modeling of full establishment. Stochastic leads to morphological variability, with size-dependent disruptions in patterning—gastruloids exceeding 500 μm often fail to sustain coordinated cell migrations, yielding fragmented rather than elongated axes. They do not incorporate systemic cues, such as maternal circulation or immune modulation, limiting insights into peri-implantation interactions and vascular integration beyond rudimentary sprouts. Metabolic perturbations, while informative, produce pleiotropic effects that confound pathway-specific causality due to the absence of compartmentalized niches. Furthermore, gastruloids halt progression around Carnegie stage 8-10 equivalents, failing to transition to without additional engineering, underscoring gaps in sustaining long-term multicellular coordination.00920-4) These constraints highlight that while gastruloids excel in dissecting molecular mechanisms, they underrepresent the biomechanical and environmental feedbacks essential for holistic embryogenesis.

Recent Advances and Future Directions

Spatiotemporal Mapping and Mechanochemical Insights

Recent advances in spatiotemporal mapping of gastrulation have integrated , single-cell RNA sequencing, and computational alignment tools to reconstruct cellular trajectories across embryonic stages. In models, a 2023 study generated a single-cell resolution spatio-temporal of germ-layer populations during gastrula stages, revealing dynamic patterns tied to positioning and . Building on this, a 2025 atlas combined spatial and temporal coordinates to map cellular diversity in embryogenesis from gastrulation onward, enabling queries into lineage progression and tissue organization. Similarly, tools like have facilitated alignment of 1.7 million s across 20 time points in embryos, reconstructing developmental trajectories with high fidelity and highlighting conserved spatiotemporal motifs in formation. These mappings underscore the precision of gastrulation, where epiblast s ingress in a temporally orchestrated manner to form and , driven by gradients of signaling molecules like and Nodal.00818-6) Human gastrulation mapping has progressed with 3D reconstructions and of intact . A 2024 analysis of a gastrulating provided cellular and molecular details of formation and specification, identifying key transcription factors such as SOX17 and GATA6 in progenitors. In 2025, of a Carnegie stage 7 resolved single-cell maps, correlating positional with modules for mesendoderm and highlighting species-specific timings relative to models. Live complemented these efforts; for instance, 2025 work described evolving mechanical properties in embryonic tissues via spatio-temporal quantification, revealing rapid transitions in cell stiffness and contractility during and . Such empirically link spatiotemporal dynamics to causal drivers, including oscillatory signaling in elongation observed through time-lapse microscopy.00357-X) Mechanochemical insights elucidate how physical forces interplay with biochemical cues to orchestrate gastrulation movements. A 2023 integrated , cortical , and chemotactic signaling to recapitulate primitive streak ingression and convergence-extension flows in chick embryos, predicting outcomes under perturbations like altered BMP gradients. This aligns with experimental evidence that feedback, such as myosin-driven contractility, coordinates epiblast cell behaviors during epithelial-to-mesenchymal transition (), a core mechanochemical process enabling . Recent 2025 modeling advances in systems further detailed formation, showing how differential and tissue viscosity generate self-organizing flows independent of global gradients, validated against live-imaging data. In luminal epithelia analogous to gastrulating tissues, mechanochemical patterning via actomyosin pulses localizes organizers, suggesting conserved mechanisms across gastrulation modes. These models emphasize causal : chemical signals initiate but properties amplify and stabilize morphogenetic patterns, with empirical perturbations confirming bidirectional feedbacks between and .

Implications for Human Development and Disorders

Disruptions in human gastrulation, which occurs during the third week of embryonic development and establishes the trilaminar germ layers and body axes, can result in early embryonic lethality or severe congenital malformations involving multiple germ layers. Abnormalities such as failed formation or defective cell migration have been linked to conditions including conjoined twinning, chordomas, and caudal dysgenesis syndromes like , where perturbations in preimplantation and gastrulation processes lead to incomplete axial elongation and fusion anomalies. , characterized by sacral agenesis, presacral masses, and anorectal malformations, exemplifies a gastrulation-related disorder tied to mutations in the MNX1 gene (formerly HLXB9), which encodes a critical for caudal development and primitive streak function. Genetic mutations affecting gastrulation regulators further underscore these implications, often manifesting as multisystem defects due to impaired organizer signaling or epithelial-mesenchymal transitions. For instance, ZIC2 mutations cause by disrupting mid-gastrulation organizer activity, leading to arrested anterior neural development and midline facial anomalies. Similarly, NIPBL in alters gastrulation-stage gene expression, phenocopying limb, craniofacial, and growth defects observed in affected individuals. Expanded HTT alleles associated with impair epiblast patterning during gastrulation, detectable as early as two weeks post-fertilization in embryonic models, potentially contributing to later neurodegenerative pathology through disrupted early cell fate decisions. These gastrulation-linked disorders highlight the process's vulnerability to genetic and environmental insults, with implications for rates—estimated at 10-20% of clinically recognized pregnancies often tracing to peri-implantation failures extending into gastrulation—and broader developmental screening. Advances in cell-derived gastruloid models enable spatiotemporal dissection of these defects, revealing mechanochemical failures that inform non-invasive prenatal diagnostics and potential regenerative interventions, though empirical limitations persist in replicating human-specific axis formation.

References

  1. [1]
    Embryology, Gastrulation - StatPearls - NCBI Bookshelf - NIH
    Apr 23, 2023 · Gastrulation is an early developmental process in which an embryo transforms from a one-dimensional layer of epithelial cells, a blastula, and reorganizes into ...Development · Cellular · Molecular Level · Function
  2. [2]
    Animal Development II: Gastrulation & Organogenesis
    Gastrulation: the dramatic rearrangement (movement) of cells in the blastula to create the embryonic tissue layers. These tissue layers will go on to produce ...
  3. [3]
    Germ Layers | Embryo Project Encyclopedia - Arizona State University
    Sep 17, 2013 · During gastrulation, a hollow cluster of cells called a blastula reorganizes into two primary germ layers: an inner layer, called endoderm, and ...
  4. [4]
    Furry is required for cell movements during gastrulation and ... - Nature
    Mar 23, 2021 · Gastrulation is a key event in animal embryogenesis during which germ layer precursors are rearranged and the embryonic axes are established ...
  5. [5]
    Embryology - Cellular Division - Gastrulation - TeachMeAnatomy
    Aug 22, 2025 · Gastrulation is a process of cellular rearrangement which involves migration, invagination and differentiation of the epiblast. It is largely ...
  6. [6]
    Embryology, Gastrulation - PubMed
    Apr 23, 2023 · Gastrulation is an early developmental process in which an embryo transforms from a one-dimensional layer of epithelial cells, a blastula, and reorganizes into ...
  7. [7]
    Gastrulation - Latest research and news - Nature
    Gastrulation is the process during which the early sphere-like embryo of most animals is dramatically re-organised by the inward migration of cells on the ...
  8. [8]
    Gastrulation - an overview | ScienceDirect Topics
    As most developmental stages, gastrulation is driven by four critical cell behaviours, i.e. cell division, cell differentiation, cell shape changes and cell ...<|separator|>
  9. [9]
    gastrulation - Wiktionary, the free dictionary
    From German Gastrulation, coined by Ernst Haeckel in his 1872 work "Biology of Calcareous Sponges" (see also gastrula).
  10. [10]
    Gastrulation in Gallus gallus (Domestic Chicken)
    Jun 21, 2011 · Ernst Haeckel coined the term; 'gaster', meaning stomach in Latin, is the root for 'gastrulation', as the gut is one of the most unique ...
  11. [11]
    Gastrula - Etymology, Origin & Meaning
    Originating in 1874 from Modern Latin, combining Latin gaster and Greek gastēr meaning "stomach," with diminutive suffix -ula; relates to gastrulation and ...
  12. [12]
    Gastrulation - UNSW Embryology
    Dec 1, 2021 · The term gastrulation means the formation of gut (Greek, gastrula = belly), but has now a more broad sense to to describe the formation of the trilaminar ...Missing: etymology | Show results with:etymology
  13. [13]
    Gastrulation: making and shaping germ layers - PubMed - NIH
    Gastrulation is a fundamental phase of animal embryogenesis during which germ layers are specified, rearranged, and shaped into a body plan with organ ...
  14. [14]
    Gastrulation: Making and Shaping Germ Layers - Annual Reviews
    Jul 9, 2012 · Gastrulation is a fundamental phase of animal embryogenesis during which germ layers are specified, rearranged, and shaped into a body plan with ...<|control11|><|separator|>
  15. [15]
    Gastrulation and the evolution of development
    Apr 1, 1992 · The origin of the gastrula can be accounted for in terms of Haeckel's gastrea theory -an early metazoan resembling the gastrula. Gastrulation in ...ABSTRACT · GASTRULATION... · THE ORIGIN OF THE... · SELECTION ON...
  16. [16]
    Establishment of the vertebrate body plan: Rethinking gastrulation ...
    Sep 13, 2021 · Conserved gene and cell regulatory networks then allow for a common body plan to be generated prior to the generation of species-specific ...
  17. [17]
    Evolutionary origin of gastrulation: insights from sponge development
    Mar 28, 2014 · The evolutionary origin of gastrulation—defined as a morphogenetic event that leads to the establishment of germ layers—remains a vexing ...Missing: etymology | Show results with:etymology
  18. [18]
    Conserved Patterns of Cell Movements during Vertebrate Gastrulation
    Current evidence indicates that the organizer activity encompasses all three germ layers of the dorsal gastrula and employs evolutionarily conserved genetic ...
  19. [19]
    Cell Movements and the Shaping of the Vertebrate Body - NCBI - NIH
    Gastrulation begins on the side of the blastula that has been marked out as dorsal by the cortical rotation. Here, involution of cells into the interior starts ...Gastrulation Transforms a... · The Movements of... · Active Changes of Cell...
  20. [20]
    Chapter 14. Gastrulation and Neurulation - Biology
    Gastrulation reorganizes the embryo into a multi-layered organism, while neurulation forms the neural tube for the spinal cord and brain.Missing: definition | Show results with:definition
  21. [21]
    [PDF] Gastrulation
    basic types of cell movements: ❖ Invagination: The infolding of a region of cells, much like the indenting of a soft rubber ball when it is poked ...
  22. [22]
    Dancing the polonaise | Nature Reviews Molecular Cell Biology
    Gastrulation is a crucial stage in embryonic development, during which extensive cell migration causes a single cell layer (ectoderm) to generate two others ( ...
  23. [23]
    3.2: Cell Division and Movement: Cleavage - Frog Gastrulation and ...
    Jan 3, 2021 · During gastrulation the cells produced during cleavage begin to move relative to each other. As they move, they change their local environment ...
  24. [24]
    [PDF] Gastrulation
    INVAGINATION -local inward buckling of an epithelium ii. INVOLUTION -inward movement of a cell layer around a point or edge iii. EPIBOLY -spread of an outside ...
  25. [25]
    [PDF] 11. Morphogenetic Movements
    Epiboly is the movement of cells on the surface of the em- brvo. Convergence, extension, etc. are epibolic movements. Emboly is the movenment of cells into ...
  26. [26]
    The evolution of gastrulation morphologies - PMC - PubMed Central
    Apr 17, 2023 · Summary: This Hypothesis reviews the mechanisms that underlie the plasticity of vertebrate gastrulation during evolution and under ...
  27. [27]
    13.2 Development and Organogenesis – Concepts of Biology
    Gastrulation leads to the formation of the three germ layers that give rise during further development to the different organs in the animal body. This process ...
  28. [28]
    Epithelial-mesenchymal transitions: the importance of changing cell ...
    This process of EMT is essential for germ layer formation and cell migration in the early vertebrate embryo (2). The process of EMT can be delineated into ...
  29. [29]
    Epithelial Mesenchymal Transition in Embryonic Development ...
    The epithelial mesenchymal transition (EMT) plays a central role in both normal physiological events (e.g., embryonic development) and abnormal pathological ...
  30. [30]
    Gastrulation and Germ Layer Formation - Creative Diagnostics
    The gastrula refers to an animal embryo with a double germ layer or a triple germ layer, an important stage in the development of animal embryos.
  31. [31]
    Back and Forth between Cell Fate Specification and Movement ...
    Clearly the major regulators of gastrulation, the Nodal, BMP, Wnt and FGF signaling pathways, can regulate both cell fates and movements. As far as Wnt ...
  32. [32]
    Reassembling gastrulation - ScienceDirect.com
    Gastrulation leads to stereotypical germ layer progenitor specification and organization. · Gastrulation relies on tight interactions between signaling pathways ...
  33. [33]
    Mouse gastrulation: coordination of tissue patterning, specification ...
    As discussed, gastrulation integrates timing and exposure to signaling pathways in order to specify cell types and pattern them prior to organogenesis.
  34. [34]
    Towards an integrated view and understanding of embryonic ...
    Apr 30, 2025 · A third signalling pathway that is required for gastrulation is Nodal signalling. Like BMP4, NODAL belongs to the TGF-β superfamily, but it ...
  35. [35]
    Setting up for gastrulation in zebrafish - PubMed
    As the basic body plan of the vertebrate embryo emerges, evolutionarily conserved developmental signaling pathways, including Bmp, Nodal, Wnt, and Fgf, direct ...
  36. [36]
    Signaling regulation during gastrulation: Insights from mouse ...
    Critical signaling pathways at gastrulation​​ WNT, BMP, Nodal and FGF signals cooperate to initiate gastrulation and germ layer specification. Disruption of any ...
  37. [37]
    Review Gastrulation Movements: the Logic and the Nuts and Bolts
    later stages of gastrulation, when the early gastrulation movements have ... The roles of three signaling pathways in the formation and function of the ...
  38. [38]
    Reconstruction of distinct vertebrate gastrulation modes via ...
    Jan 4, 2023 · Manipulation of mesendoderm territory and cell behavior recreates distinct vertebrate gastrulation morphologies in the chick.
  39. [39]
    From blastocyst to gastrula: gene regulatory networks of embryonic ...
    Here, we review how regulatory networks constructed for different stem cell types relate to corresponding networks in vivo and provide insights into ...
  40. [40]
    GATA4/5/6 family transcription factors are conserved determinants of ...
    Our work uncovers the highly dynamic, lineage-specific transcriptional regulation of gata5/6 in multiple mesendoderm lineages during zebrafish gastrulation.
  41. [41]
    Interspecies control of development during mammalian gastrulation
    Nov 7, 2023 · The authors identified a highly conserved 'regulatory core' of 75 transcription factors that regulate cell states. Furthermore, there was a ...
  42. [42]
    The primitive streak and cellular principles of building an amniote ...
    Dec 3, 2021 · Genetic studies have identified a conserved network of signaling molecules and transcription factors associated with gastrulation and have ...
  43. [43]
    Gastrulation in Drosophila melanogaster: Genetic control, cellular ...
    This review covers topics ranging from the identification and functional characterization of developmental and morphogenetic control genes to the analysis of ...
  44. [44]
    Epigenetic control of cell identities from epiblast to gastrulation
    Feb 22, 2025 · In this review, we cover the contribution of dynamic chromatin modifications by DNA methylation, changes of chromatin accessibility, and histone modifications.<|separator|>
  45. [45]
    Epigenetic Control of Cell Potency and Fate Determination during ...
    May 25, 2023 · Embryonic Stem Cells Are a Useful Model to Study Epigenetic Regulation during Gastrulation. Epigenetic regulation of gene expression is critical ...
  46. [46]
    [PDF] Epigenetic regulator function through mouse gastrulation
    We examined changes at CpG islands (CGIs), which represent a major focal point of Polycomb and Trithorax group-based regulation and are usually free of DNA ...
  47. [47]
    The interplay between DNA and histone methylation - EMBO Press
    Apr 12, 2021 · A strong correlation exists between the genome‐wide distribution of DNA and histone methylation, suggesting an intimate relationship between these epigenetic ...<|control11|><|separator|>
  48. [48]
    Single-cell multi-omics delineates the dynamics of distinct ...
    May 8, 2025 · Our study broadens the current understanding of intricate epigenetic regulatory networks governing mouse gastrulation and sheds light on their relevance to ...
  49. [49]
    Integrated single-cell sequencing reveals principles of epigenetic ...
    Integrated single-cell sequencing reveals principles of epigenetic regulation of human gastrulation and germ cell development in a 3D organoid model.
  50. [50]
    Epigenetic profiling and incidence of disrupted development point to ...
    Aug 4, 2022 · Our data point to gastrulation as a critical stage for aging and natural rejuvenation, characterized by the lowest epigenetic age, increased mortality.
  51. [51]
    Section 2: Gastrulation in Protostomes and Deuterostomes
    Gastrulation is a transformative phase in early embryonic development, where the embryo progresses from a simple, spherical blastula into a multi-layered ...
  52. [52]
    Features Used to Classify Animals - OpenEd CUNY
    One of these differences between protostomes and deuterostomes is the method of coelom formation, beginning from the gastrula stage. Since body cavity formation ...
  53. [53]
    Sea Urchin as a Universal Model for Studies of Gene Networks - NIH
    Jan 20, 2021 · The purple sea urchin Strongylocentrotus purpuratus has been used for over 150 years as a model organism in developmental biology.
  54. [54]
    [PDF] Sea Urchins as a Model System for Studying Embryonic Development
    Sea urchins are deuterostomes; they gastrulate through a blastopore which becomes the anus, while the mouth forms later as a secondary opening between the ...
  55. [55]
    Gastrulation in the sea urchin - PMC - NIH
    Gastrulation in the sea urchin is a beautifully orchestrated sequence of morphogenetic movements. After over a century of study the molecular underpinnings of ...
  56. [56]
    Growth factor-mediated mesodermal cell guidance and ...
    Oct 15, 2013 · In this study, we aimed to further elucidate the roles of VEGF and FGF signaling in gastrulation and skeletogenesis in the sea urchin. We ...Vegf And Fgf Ligands Are... · Vegf Signaling Plays A More... · Vegf Signaling Controls Pmc...
  57. [57]
    The gene regulatory control of sea urchin gastrulation - ScienceDirect
    This review discusses the major cell movements that take place during sea urchin gastrulation with an emphasis on the gene regulatory basis of those movements.
  58. [58]
    [PDF] The gene regulatory control of sea urchin gastrulation
    More comprehensive reviews of various aspects of sea urchin gastrulation are available (Hardin, 1996; Kominami and Takata, 2004;. McClay, 2004). 2. General ...
  59. [59]
    Gastrulation in Xenopus | Embryo Project Encyclopedia
    Mar 10, 2011 · The process of gastrulation allows for the formation of the germ layers in metazoan embryos, and is generally achieved through a series of ...
  60. [60]
    Early Amphibian Development - Developmental Biology - NCBI - NIH
    Before we look at the process of gastrulation in detail, let us first trace the movement patterns of the germ layers. Gastrulation in frog embryos is initiated ...
  61. [61]
    Mechanism of Gastrulation In Amphibians: A Comprehensive Review
    In amphibians, gastrulation begins with the formation of a blastopore, which marks the beginning of the primitive gut.
  62. [62]
    Convergent Extension in the Amphibian, Xenopus laevis - PMC
    Convergence force rises at the onset of gastrulation to 1.5-2.0 μN when it plateaus through the end of gastrulation, and then rises to over 4.0 μN by the ...
  63. [63]
    Cell migration in the Xenopus gastrula - Huang - 2018
    Jun 26, 2018 · Xenopus gastrulation movements are in large part based on the rearrangement of cells by differential cell-on-cell migration within ...
  64. [64]
    Molecular Nature of Spemann's Organizer: the Role of the Xenopus ...
    This study analyzes the function of the homeobox gene goosecoid in Xenopus development. First, we find that goosecoid mRNA distribution closely mimics the ...
  65. [65]
    Induction into the Hall of Fame: tracing the lineage of Spemann's ...
    Oct 15, 2008 · In their 1983 study, Smith and Slack decided to repeat the organizer graft experiments of Spemann and Mangold in Xenopus laevis, rather than in ...
  66. [66]
    Chapter 5 Cell Adhesion in Amphibian Gastrulation - ScienceDirect
    It consists of the establishment of the germ layers by a rearrangement of embryonic parts in which the future endoderm, and mesoderm if present, move from the ...
  67. [67]
    The function and mechanism of convergent extension during ...
    Nov 1, 1985 · The processes thought to function in Xenopus gastrulation include bottle cell formation, migration of cells on the roof of the blastocoel, ...
  68. [68]
  69. [69]
    Molecular anatomy of the pre-primitive-streak chick embryo - Journals
    Feb 12, 2020 · After laying, about 15 h elapse before the beginning of gastrulation, marked by the appearance of the primitive streak, the site of mesoderm and ...Introduction · Terminology and staging · Results · Discussion
  70. [70]
    Induction and patterning of the primitive streak, an organizing center ...
    Jan 28, 2004 · The primitive streak is the organizing center for amniote gastrulation. It defines the future embryonic midline and serves as a conduit of ...
  71. [71]
    Christian Heinrich Pander (1794-1865) - Wiley Online Library
    This major discovery, which helped focus research into subsequent morphogenesis of these layers, is usually attributed to the work of Christian Pander in 1817, ...
  72. [72]
    [PDF] Amphibian gastrulation: history and evolution of a 125 year-old ...
    1911 Goodale First use of vital dye staining on an Amphibian blastula. 1923 a,b Vogt First comprehensive drawings of vital dye mark movements. Mesoderm origin ...
  73. [73]
    Spemann's organizer and self-regulation in amphibian embryos
    Feb 1, 2006 · In 1924, Spemann and Mangold demonstrated the induction of Siamese twins in transplantation experiments with salamander eggs.
  74. [74]
    Amphibian Gastrulation: History and Evolution of a 125 Year-Old ...
    Holtfreter (1943,1944) illuminated unknown aspects of living gastrula cells and his observations helped to define many problems to be solved. During the second ...
  75. [75]
    Origin and evolution of endoderm and mesoderm - PubMed
    The evolutionary origin of the inner germ layers, endoderm and mesoderm, and their relationship have been a matter of debate for decades.
  76. [76]
    Cnidarian-bilaterian comparison reveals the ancestral regulatory ...
    Jun 29, 2021 · As a bilaterian sister group, cnidarians provide us with a key reference point regarding the evolution of body axes patterning and germ layer ...
  77. [77]
    Resolving homology in the face of shifting germ layer origins - eLife
    Dec 23, 2019 · These concepts provide a general framework for approaching issues of homology in cases where germ layer origins have shifted during evolution.
  78. [78]
    Molecular evidence from xenacoelomorph gonopore formation supports homology with the bilaterian anus - Nature Ecology & Evolution
    ### Summary of Findings on Gut-Like Ectodermal Tissue in Sea Anemone and Implications for Bilaterian Anus Homology
  79. [79]
    The evolution of gastrulation morphologies | Development
    Apr 17, 2023 · ... germ layers ... L. and. Arias. ,. A. M.. (. 2021. ). Establishment of the vertebrate body plan: Rethinking gastrulation through stem cell models ...
  80. [80]
    Interspecies control of development during mammalian gastrulation
    Nov 7, 2023 · We provide an overview of the latest research on interspecies developmental control across mammals. This includes single-cell atlases of several mammalian ...
  81. [81]
    Spatiotemporal transcriptomics reveals the evolutionary history of ...
    Mar 12, 2015 · Of the three germ layers, the mesoderm is found in bilaterian animals but is absent in species in the phyla Cnidaria and Ctenophora, which has ...Missing: organisms | Show results with:organisms
  82. [82]
    Gastruloids as in vitro models of embryonic blood development with ...
    Aug 4, 2022 · In conclusion, our data suggest that gastruloids are excellent candidates to model hematopoietic development in vitro in an embryo-like context.
  83. [83]
    [PDF] Generating gastruloids with somite-like structures from mouse ...
    - Make sure that DNA LoBind tips instead of regular tips are used to add the cells or medium to the 96-well plate. Gastruloids do not elongate. - Make sure to ...
  84. [84]
    A method to recapitulate early embryonic spatial patterning in ...
    Gastrulation-like events in micropatterned differentiation. The expression of BRA and the emergence of all three germ layers in the micropatterned cultures ...
  85. [85]
    Generating Gastruloids from Mouse Embryonic Stem Cells
    Oct 12, 2018 · Here, we present a 5-7 day protocol for generating aggregates of mESCs, that we have dubbed gastruloids, that mimic the early post-implantation ...
  86. [86]
    Gastruloids: Pluripotent stem cell models of mammalian gastrulation ...
    Gastruloids are a models of early mammalian embryogenesis based on Embryonic Stem (ES) cells. Gastruloids derived from mouse and human reveal a hereto unknown ...
  87. [87]
    Gastruloids: Pluripotent stem cell models of mammalian gastrulation ...
    Gastruloids are 3D aggregates of stem cells that model gastrulation, showing symmetry breaking and germ layer specification, but lack brain structures.
  88. [88]
    A New Approach to Generate Gastruloids to Develop Anterior ...
    Jul 20, 2023 · We report a novel culture protocol to coax mESCs into post-implantation epiblast-like (EPI) aggregates in high throughput on bioengineered microwell arrays.
  89. [89]
    Advances in engineered models of peri-gastrulation - PMC
    The formation of gastruloids in vitro involves the activation of three crucial cell signaling pathways: Wingless Int-1 (WNT), bone morphogenic protein (BMP), ...
  90. [90]
    Chimeric 3D gastruloids – a versatile tool for studies of mammalian ...
    Chimeric gastruloids can be used as versatile novel model system for various types of studies and embryonic research questions, as indicated. (B) Schematic of ...
  91. [91]
    3D gastruloids: a novel frontier in stem cell-based in vitro modeling ...
    3D gastruloids provide an unparalleled opportunity to study normal and pathological embryogenesis from a bottom-up perspective and in a high-throughput manner.
  92. [92]
    Gastruloids — a minimalistic model to study complex developmental ...
    Dec 18, 2023 · Together, we propose that gastruloids are an ideal system to rapidly uncover new mechanistic links between developmental signalling pathways and ...
  93. [93]
    Retinoic acid induces human gastruloids with posterior embryo-like ...
    Aug 20, 2024 · Here we show that an early pulse of retinoic acid (RA), together with later Matrigel, robustly induces human gastruloids with posterior embryo-like ...Missing: achievements | Show results with:achievements
  94. [94]
  95. [95]
    Gastruloids enable modeling of the earliest stages of human cardiac ...
    Jun 5, 2025 · We developed an in vitro model to mimic the earliest developmental stages of cardiac and hepatic organoid vascularization.Missing: achievements | Show results with:achievements
  96. [96]
    Gastruloids are competent to specify both cardiac and skeletal ...
    Nov 23, 2024 · In this work, we unveil the potential of gastruloids to undergo specification into both cardiac and skeletal muscle lineages.Missing: achievements | Show results with:achievements
  97. [97]
    Development of large-scale gastruloid array to identify aberrant ...
    Jun 10, 2025 · We developed a microraft array-based technology to perform image-based assays of large numbers of fixed or living gastruloids and sort individual gastruloids ...
  98. [98]
  99. [99]
    Morphogenetic constrains in the development of gastruloids - bioRxiv
    Dec 20, 2024 · We report that gastruloids exhibit robust morphology and transcriptional profiles within a size range. However, size affects the dynamics, and, ...Missing: achievements limitations
  100. [100]
    Time space and single-cell resolved tissue lineage trajectories and ...
    Sep 14, 2023 · Time space and single-cell resolved tissue lineage trajectories and laterality of body plan at gastrulation ... germ layers during gastrulation.
  101. [101]
    Mapping cells through time and space with moscot - Nature
    Jan 22, 2025 · We demonstrate the capability of moscot to efficiently reconstruct developmental trajectories of 1.7 million cells from mouse embryos across 20 time points.
  102. [102]
    Spatial transcriptomic characterization of a Carnegie stage 7 human ...
    Jan 10, 2025 · Here we show the use of spatial transcriptomics to analyse a fully intact Carnegie stage 7 human embryo at single-cell resolution, along with ...
  103. [103]
    Highly dynamic mechanical transitions in embryonic cell populations ...
    Jul 14, 2025 · The first major, embryo-scale, morphogenetic event in metazoans is gastrulation, which connects cell fate specification with the coordinated ...
  104. [104]
    A mechanochemical model recapitulates distinct vertebrate ...
    Dec 6, 2023 · During vertebrate gastrulation, an embryo transforms from a layer of epithelial cells into a multilayered gastrula.
  105. [105]
    Advances in mechanochemical modelling of vertebrate gastrulation
    Jul 21, 2025 · We outline advances in modelling the mechanochemical processes controlling primitive streak formation in the early avian embryo and discuss ...
  106. [106]
    Mechanochemical patterning localizes the organizer of a luminal ...
    Jun 25, 2025 · This work highlights a versatile mechanochemical mechanism for luminal epithelium patterning, which is relevant across various biological systems.
  107. [107]
    Current Concepts and Implications for Spinal Malformations - PMC
    There are a number of human malformations and disorders whose etiology has been attributed to abnormal gastrulation, these include conjoined twinning, chordomas ...
  108. [108]
    Birth Defects Associated with Perturbations in Pre-implantation ...
    Moreover, even though we will discuss caudal dysgenesis and sirenomelia as congenital defects occuring during gastrulation and axial elongation, their etiology ...
  109. [109]
    Zic2 -associated holoprosencephaly is caused by a transient defect ...
    Mutation of Zic2 causes HPE via a transient defect in the function of the organizer region at mid-gastrulation which causes an arrest in the development of the ...Results · Shh And Zic2 Do Not Interact... · Shh Signalling In The...<|control11|><|separator|>
  110. [110]
    Gastrulation-stage gene expression in Nipbl +/− mouse embryos ...
    Nipbl deficiency phenocopies gene expression changes and birth defects seen in Cornelia de Lange syndrome, the most common cause of which is Nipbl ...
  111. [111]
    HTT Mutations Affect Embryo Development as Early as 2 Weeks
    Mar 15, 2022 · Huntington's disease-causing mutations in the HTT gene lead to abnormalities in embryonic development soon after conception, a study says.
  112. [112]
    Implantation and Gastrulation Abnormalities Characterize Early ...
    Oct 8, 2020 · Mutant embryos can be classified into 5 categories: abnormal gastrulation, absence of primitive streak, developmental delay, extra-embryonic ...
  113. [113]
    Human gastrulation: The embryo and its models - ScienceDirect.com
    Here we summarize the cellular, molecular, and temporal aspects associated with human gastrulation and discuss how they relate to existing human PSCs based ...