Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 3.625
Filter
1.
Biol Open ; 13(8)2024 Aug 15.
Article in English | MEDLINE | ID: mdl-39162010

ABSTRACT

Collectively migrating Xenopus mesendoderm cells are arranged into leader and follower rows with distinct adhesive properties and protrusive behaviors. In vivo, leading row mesendoderm cells extend polarized protrusions and migrate along a fibronectin matrix assembled by blastocoel roof cells. Traction stresses generated at the leading row result in the pulling forward of attached follower row cells. Mesendoderm explants removed from embryos provide an experimentally tractable system for characterizing collective cell movements and behaviors, yet the cellular mechanisms responsible for this mode of migration remain elusive. We introduce a novel agent-based computational model of migrating mesendoderm in the Cellular-Potts computational framework to investigate the respective contributions of multiple parameters specific to the behaviors of leader and follower row cells. Sensitivity analyses identify cohesotaxis, tissue geometry, and cell intercalation as key parameters affecting the migration velocity of collectively migrating cells. The model predicts that cohesotaxis and tissue geometry in combination promote cooperative migration of leader cells resulting in increased migration velocity of the collective. Radial intercalation of cells towards the substrate is an additional mechanism contributing to an increase in migratory speed of the tissue. Model outcomes are validated experimentally using mesendoderm tissue explants.


Subject(s)
Cell Movement , Models, Biological , Xenopus , Animals , Xenopus/embryology , Mesoderm/cytology , Mesoderm/embryology , Cell Adhesion , Xenopus laevis/embryology , Computer Simulation
2.
Mol Brain ; 17(1): 58, 2024 Aug 22.
Article in English | MEDLINE | ID: mdl-39175067

ABSTRACT

Protocadherin 19 (Pcdh19) is a homophilic cell adhesion molecule and is involved in a variety of neuronal functions. Here, we tested whether Pcdh19 has a regulatory role in axon guidance using the developing Xenopus retinotectal system. We performed targeted microinjections of a translation blocking antisense morpholino oligonucleotide to knock down the expression of Pcdh19 selectively in the central nervous system. Knocking down Pcdh19 expression resulted in navigational errors of retinal ganglion cell (RGC) axons specifically at the optic chiasm. Instead of projecting to the contralateral optic tectum, RGC axons in the Pcdh19-depleted embryo misprojected ipsilaterally. Although incorrectly delivered into the ipsilateral brain hemisphere, these axons correctly reached the optic tectum. These data suggest that Pcdh19 has a critical role in preventing mixing of RGC axons originating from the opposite eyes at the optic chiasm, highlighting the importance of cell adhesion in bundling of RGC axons.


Subject(s)
Axon Guidance , Axons , Cadherins , Protocadherins , Retinal Ganglion Cells , Xenopus Proteins , Xenopus laevis , Animals , Cadherins/metabolism , Xenopus Proteins/metabolism , Xenopus Proteins/genetics , Retinal Ganglion Cells/metabolism , Xenopus laevis/embryology , Axons/metabolism , Retina/metabolism , Retina/embryology , Visual Pathways , Gene Knockdown Techniques , Optic Chiasm/embryology , Optic Chiasm/metabolism , Superior Colliculi/embryology , Superior Colliculi/metabolism , Gene Expression Regulation, Developmental
3.
Cells ; 13(16)2024 Aug 21.
Article in English | MEDLINE | ID: mdl-39195278

ABSTRACT

Retinal progenitor cells (RPCs) are a multipotent and highly proliferative population that give rise to all retinal cell types during organogenesis. Defining their molecular signature is a key step towards identifying suitable approaches to treat visual impairments. Here, we performed RNA sequencing of whole eyes from Xenopus at three embryonic stages and used differential expression analysis to define the transcriptomic profiles of optic tissues containing proliferating and differentiating RPCs during retinogenesis. Gene Ontology and KEGG pathway analyses showed that genes associated with developmental pathways (including Wnt and Hedgehog signaling) were upregulated during the period of active RPC proliferation in early retinal development (Nieuwkoop Faber st. 24 and 27). Developing eyes had dynamic expression profiles and shifted to enrichment for metabolic processes and phototransduction during RPC progeny specification and differentiation (st. 35). Furthermore, conserved adult eye regeneration genes were also expressed during early retinal development, including sox2, pax6, nrl, and Notch signaling components. The eye transcriptomic profiles presented here span RPC proliferation to retinogenesis and include regrowth-competent stages. Thus, our dataset provides a rich resource to uncover molecular regulators of RPC activity and will allow future studies to address regulators of RPC proliferation during eye repair and regrowth.


Subject(s)
Gene Expression Profiling , Gene Expression Regulation, Developmental , Transcriptome , Xenopus laevis , Animals , Xenopus laevis/genetics , Xenopus laevis/embryology , Transcriptome/genetics , Eye/metabolism , Eye/embryology , Retina/metabolism , Retina/growth & development , Cell Differentiation/genetics , Cell Proliferation/genetics , Organogenesis/genetics , Stem Cells/metabolism , Stem Cells/cytology , Xenopus Proteins/genetics , Xenopus Proteins/metabolism , Signal Transduction/genetics
4.
Dev Biol ; 515: 46-58, 2024 Nov.
Article in English | MEDLINE | ID: mdl-38968989

ABSTRACT

The Daam1 protein regulates Wnt-induced cytoskeletal changes during vertebrate gastrulation though its full mode of action and binding partners remain unresolved. Here we identify Reversion Induced LIM domain protein (RIL) as a new interacting protein of Daam1. Interaction studies uncover binding of RIL to the C-terminal actin-nucleating portion of Daam1 in a Wnt-responsive manner. Immunofluorescence studies showed subcellular localization of RIL to actin fibers and co-localization with Daam1 at the plasma membrane. RIL gain- and loss-of-function approaches in Xenopus produced severe gastrulation defects in injected embryos. Additionally, a simultaneous loss of Daam1 and RIL synergized to produce severe gastrulation defects indicating RIL and Daam1 may function in the same signaling pathway. RIL further synergizes with another novel Daam1-interacting protein, Formin Binding Protein 1 (FNBP1), to regulate gastrulation. Our studies altogether show RIL mediates Daam1-regulated non-canonical Wnt signaling that is required for vertebrate gastrulation.


Subject(s)
Actin Cytoskeleton , Gastrulation , Microfilament Proteins , Wnt Signaling Pathway , Xenopus Proteins , Xenopus laevis , Animals , Female , Humans , Rats , Actin Cytoskeleton/metabolism , Actins/metabolism , Adaptor Proteins, Signal Transducing , LIM Domain Proteins/metabolism , LIM Domain Proteins/genetics , Microfilament Proteins/metabolism , Microfilament Proteins/genetics , Protein Binding , Wnt Signaling Pathway/physiology , Xenopus laevis/embryology , Xenopus laevis/metabolism , Xenopus Proteins/metabolism , Xenopus Proteins/genetics
5.
Reprod Toxicol ; 128: 108632, 2024 09.
Article in English | MEDLINE | ID: mdl-38971262

ABSTRACT

The aim of the present work is to propose a new quantitative assessment method (FETAX-score) for determining the degree of Xenopus laevis embryo development intended for use in embryotoxicity studies. Inspired by a similar scoring system used to evaluate developmental delays (young-for-age phenotypes) in rat embryos cultured in vitro, the FETAX-score was established by considering seven morphological features (head, naris, mouth, lower jaw, tentacles, intestine, anus) that are easily evaluable in tadpoles during the late stages of development at the conclusion of the test. Given that X. laevis development is temperature-dependent and that temperatures below 14°C and above 26°C are teratogenic, the FETAX-score was tested in embryos maintained at 17, 20, 23 and 26°C. No abnormalities were observed in any group, while the total score was temperature-related, suggesting that the FETAX-score is sensitive to moderate distress that does not influence general morphology. Intestine and anus were the least sensitive structures to temperature variations. To assess the applicability of the FETAX-score in developmental toxicological studies, we evaluated FETAX-score in tadpoles exposed during the morphogenetic period to Ethanol (Eth) at concentrations of 0, 0.25, 0.5, 1, 1.5, and 2 % v/v. Gross malformations were observed only in tadpoles from the Eth 2 % group. By contrast, data analysis of the other Eth groups showed dose-related reductions in the FETAX-score. Tentacles were the most sensitive structures to Eth-related delays. These results support the use of the FETAX-score to quantitatively assess developmental deviations in FETAX embryotoxicity studies.


Subject(s)
Embryo, Nonmammalian , Embryonic Development , Ethanol , Temperature , Teratogens , Xenopus laevis , Animals , Xenopus laevis/embryology , Ethanol/toxicity , Embryo, Nonmammalian/drug effects , Embryo, Nonmammalian/abnormalities , Embryonic Development/drug effects , Teratogens/toxicity , Larva/drug effects , Larva/growth & development , Toxicity Tests/methods , Abnormalities, Drug-Induced
6.
Proc Natl Acad Sci U S A ; 121(28): e2408346121, 2024 Jul 09.
Article in English | MEDLINE | ID: mdl-38968117

ABSTRACT

Xenopus embryos provide a favorable material to dissect the sequential steps that lead to dorsal-ventral (D-V) and anterior-posterior (A-P) cell differentiation. Here, we analyze the signaling pathways involved in this process using loss-of-function and gain-of-function approaches. The initial step was provided by Hwa, a transmembrane protein that robustly activates early ß-catenin signaling when microinjected into the ventral side of the embryo leading to complete twinned axes. The following step was the activation of Xenopus Nodal-related growth factors, which could rescue the depletion of ß-catenin and were themselves blocked by the extracellular Nodal antagonists Cerberus-Short and Lefty. During gastrulation, the Spemann-Mangold organizer secretes a cocktail of growth factor antagonists, of which the BMP antagonists Chordin and Noggin could rescue simultaneously D-V and A-P tissues in ß-catenin-depleted embryos. Surprisingly, this rescue occurred in the absence of any ß-catenin transcriptional activity as measured by ß-catenin activated Luciferase reporters. The Wnt antagonist Dickkopf (Dkk1) strongly synergized with the early Hwa signal by inhibiting late Wnt signals. Depletion of Sizzled (Szl), an antagonist of the Tolloid chordinase, was epistatic over the Hwa and Dkk1 synergy. BMP4 mRNA injection blocked Hwa-induced ectopic axes, and Dkk1 inhibited BMP signaling late, but not early, during gastrulation. Several unexpected findings were made, e.g., well-patterned complete embryonic axes are induced by Chordin or Nodal in ß-catenin knockdown embryos, dorsalization by Lithium chloride (LiCl) is mediated by Nodals, Dkk1 exerts its anteriorizing and dorsalizing effects by regulating late BMP signaling, and the Dkk1 phenotype requires Szl.


Subject(s)
Body Patterning , Intercellular Signaling Peptides and Proteins , Signal Transduction , Xenopus Proteins , beta Catenin , Animals , Body Patterning/genetics , Xenopus Proteins/metabolism , Xenopus Proteins/genetics , beta Catenin/metabolism , beta Catenin/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Intercellular Signaling Peptides and Proteins/genetics , Xenopus laevis/embryology , Gene Expression Regulation, Developmental , Gastrulation , Nodal Protein/metabolism , Nodal Protein/genetics , Embryo, Nonmammalian/metabolism , Embryo, Nonmammalian/embryology , Organizers, Embryonic/metabolism , Glycoproteins
7.
Int J Dev Biol ; 68(2): 85-91, 2024 Jul 15.
Article in English | MEDLINE | ID: mdl-39016375

ABSTRACT

The tRNA-histidine guanylyltransferase 1-like (THG1L), also known as induced in high glucose-1 (IHG-1), encodes for an essential mitochondria-associated protein highly conserved throughout evolution, that catalyses the 3'-5' addition of a guanine to the 5'-end of tRNA-histidine (tRNAHis). Previous data indicated that THG1L plays a crucial role in the regulation of mitochondrial biogenesis and dynamics, in ATP production, and is critically involved in the modulation of apoptosis, cell-cycle progression and survival, as well as in cellular stress responses and redox homeostasis. Dysregulations of THG1L expression play a central role in various pathologies, including nephropathies, and neurodevelopmental disorders often characterized by developmental delay and cerebellar ataxia. Despite the essential role of THG1L, little is known about its expression during vertebrate development. Herein, we examined the detailed spatio-temporal expression of this gene in the developing Xenopus laevis. Our results show that thg1l is maternally inherited and its temporal expression suggests a role during the earliest stages of embryogenesis. Spatially, thg1l mRNA localizes in the ectoderm and marginal zone mesoderm during early stages of development. Then, at tadpole stages, thg1l transcripts mostly localise in neural crests and their derivatives, somites, developing kidney and central nervous system, therefore largely coinciding with territories displaying intense energy metabolism during organogenesis in Xenopus.


Subject(s)
Gene Expression Regulation, Developmental , Nucleotidyltransferases , Xenopus Proteins , Xenopus laevis , Animals , Embryo, Nonmammalian/metabolism , Embryo, Nonmammalian/embryology , Embryonic Development/genetics , Nucleotidyltransferases/genetics , Nucleotidyltransferases/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Xenopus laevis/metabolism , Xenopus laevis/embryology , Xenopus laevis/genetics , Xenopus Proteins/genetics , Xenopus Proteins/metabolism
8.
EMBO Rep ; 25(8): 3300-3323, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38943003

ABSTRACT

Heterochromatin marks such as H3K9me3 undergo global erasure and re-establishment after fertilization, and the proper reprogramming of H3K9me3 is essential for early development. Despite the widely conserved dynamics of heterochromatin reprogramming in invertebrates and non-mammalian vertebrates, previous studies have shown that the underlying mechanisms may differ between species. Here, we investigate the molecular mechanism of H3K9me3 dynamics in medaka (Japanese killifish, Oryzias latipes) as a non-mammalian vertebrate model, and show that rapid cell cycle during cleavage stages causes DNA replication-dependent passive erasure of H3K9me3. We also find that cell cycle slowing, toward the mid-blastula transition, permits increasing nuclear accumulation of H3K9me3 histone methyltransferase Setdb1, leading to the onset of H3K9me3 re-accumulation. We further demonstrate that cell cycle length in early development also governs H3K9me3 reprogramming in zebrafish and Xenopus laevis. Together with the previous studies in invertebrates, we propose that a cell cycle length-dependent mechanism for both global erasure and re-accumulation of H3K9me3 is conserved among rapid-cleavage species of non-mammalian vertebrates and invertebrates such as Drosophila, C. elegans, Xenopus and teleost fish.


Subject(s)
Cell Cycle , Heterochromatin , Histones , Oryzias , Animals , Heterochromatin/metabolism , Heterochromatin/genetics , Histones/metabolism , Cell Cycle/genetics , Oryzias/embryology , Oryzias/genetics , Oryzias/metabolism , Zebrafish/genetics , Zebrafish/embryology , Xenopus laevis/embryology , Xenopus laevis/metabolism , DNA Replication , Cellular Reprogramming/genetics , Histone-Lysine N-Methyltransferase/metabolism , Histone-Lysine N-Methyltransferase/genetics
9.
Dev Biol ; 515: 18-29, 2024 Nov.
Article in English | MEDLINE | ID: mdl-38945423

ABSTRACT

The Formin protein Daam1 is required for Wnt-induced cytoskeletal changes during gastrulation, though how it accomplishes this remains unresolved. Here we report the characterization of Formin Binding Protein 1 (FNBP1) as a binding partner of Daam1. The interaction of Daam1 with FNBP1 and its domains required for this interaction were delineated. Immunofluorescence studies showed FNBP1 co-localizes with Daam1, and is an integral component of the actin cytoskeletal complex that is responsive to Wnt stimulation. Specifically, FNBP1 can induce intracellular tubule-like structures and localize to focal adhesions suggesting a role for FNBP1 in cell migration. Functional FNBP1 studies in Xenopus embryos uncover a critical role for FNBP1 in regulating vertebrate gastrulation. Additionally, suboptimal doses of Daam1 and FNBP1 synergize to produce severe gastrulation defects, indicating FNBP1 and Daam1 may function within the same signaling pathway. These results together show FNBP1 is an integral component of Daam1-regulated non-canonical Wnt signaling required for vertebrate gastrulation.


Subject(s)
Gastrulation , Wnt Signaling Pathway , Xenopus Proteins , Xenopus laevis , Animals , Humans , Mice , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/genetics , Cell Movement , Embryo, Nonmammalian/metabolism , Microfilament Proteins/metabolism , Microfilament Proteins/genetics , Protein Binding , rho GTP-Binding Proteins , Wnt Signaling Pathway/physiology , Xenopus laevis/embryology , Xenopus laevis/metabolism , Xenopus Proteins/metabolism , Xenopus Proteins/genetics , Female
10.
Development ; 151(13)2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38884356

ABSTRACT

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


Subject(s)
Neural Crest , Animals , Neural Crest/cytology , Neural Crest/metabolism , Epigenesis, Genetic , Gene Expression Regulation, Developmental , Xenopus Proteins/metabolism , Xenopus Proteins/genetics , Xenopus laevis/embryology , Blastula/metabolism , Blastula/cytology , Cell Differentiation , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Transcriptome/genetics
11.
Dev Biol ; 514: 28-36, 2024 Oct.
Article in English | MEDLINE | ID: mdl-38880277

ABSTRACT

Primordial germ cells (PGCs) are the precursors of sperms and oocytes. Proper development of PGCs is crucial for the survival of the species. In many organisms, factors responsible for PGC development are synthesized during early oogenesis and assembled into the germ plasm. During early embryonic development, germ plasm is inherited by a few cells, leading to the formation of PGCs. While germline development has been extensively studied, how components of the germ plasm regulate PGC development is not fully understood. Here, we report that Dzip1 is dynamically expressed in vertebrate germline and is a novel component of the germ plasm in Xenopus and zebrafish. Knockdown of Dzip1 impairs PGC development in Xenopus embryos. At the molecular level, Dzip1 physically interacts with Dazl, an evolutionarily conserved RNA-binding protein that plays a multifaced role during germline development. We further showed that the sequence between amino acid residues 282 and 550 of Dzip1 is responsible for binding to Dazl. Disruption of the binding between Dzip1 and Dazl leads to defective PGC development. Taken together, our results presented here demonstrate that Dzip1 is dynamically expressed in the vertebrate germline and plays a novel function during Xenopus PGC development.


Subject(s)
Adaptor Proteins, Signal Transducing , Gene Expression Regulation, Developmental , Germ Cells , RNA-Binding Proteins , Xenopus Proteins , Xenopus laevis , Animals , Female , Germ Cells/metabolism , Germ Cells/cytology , Oogenesis/genetics , RNA-Binding Proteins/metabolism , RNA-Binding Proteins/genetics , Xenopus laevis/embryology , Xenopus laevis/metabolism , Xenopus laevis/genetics , Xenopus Proteins/metabolism , Xenopus Proteins/genetics , Zebrafish/embryology , Zebrafish/genetics , Zebrafish/metabolism , Zebrafish Proteins/metabolism , Zebrafish Proteins/genetics , Adaptor Proteins, Signal Transducing/metabolism
12.
Mol Cells ; 47(6): 100076, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38825188

ABSTRACT

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


Subject(s)
Cell Movement , Destrin , Embryonic Development , Xenopus laevis , Animals , Xenopus laevis/embryology , Xenopus laevis/metabolism , Destrin/metabolism , Destrin/genetics , Xenopus Proteins/metabolism , Xenopus Proteins/genetics , Neural Crest/metabolism , Neural Crest/embryology , Neural Crest/cytology , Neurogenesis , Neural Plate/metabolism , Neural Plate/embryology , Actins/metabolism , Gene Expression Regulation, Developmental
13.
Dev Growth Differ ; 66(5): 320-328, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38925637

ABSTRACT

During the formation of the neural tube, the primordium of the vertebrate central nervous system, the actomyosin activity of cells in different regions drives neural plate bending. However, how the stiffness of the neural plate and surrounding tissues is regulated and mechanically influences neural plate bending has not been elucidated. Here, we used atomic force microscopy to reveal the relationship between the stiffness of the neural plate and the mesoderm during Xenopus neural tube formation. Measurements with intact embryos revealed that the stiffness of the neural plate was consistently higher compared with the non-neural ectoderm and that it increased in an actomyosin activity-dependent manner during neural plate bending. Interestingly, measurements of isolated tissue explants also revealed that the relationship between the stiffness of the apical and basal sides of the neural plate was reversed during bending and that the stiffness of the mesoderm was lower than that of the basal side of the neural plate. The experimental elevation of mesoderm stiffness delayed neural plate bending, suggesting that low mesoderm stiffness mechanically supports neural tube closure. This study provides an example of mechanical interactions between tissues during large-scale morphogenetic movements.


Subject(s)
Neural Plate , Neural Tube , Xenopus laevis , Animals , Neural Tube/embryology , Neural Tube/cytology , Neural Tube/metabolism , Neural Plate/embryology , Neural Plate/metabolism , Neural Plate/cytology , Xenopus laevis/embryology , Mesoderm/cytology , Mesoderm/embryology , Mesoderm/metabolism , Ectoderm/cytology , Ectoderm/metabolism , Microscopy, Atomic Force , Embryo, Nonmammalian/cytology , Embryo, Nonmammalian/metabolism , Embryo, Nonmammalian/embryology
14.
Dev Biol ; 512: 26-34, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38705558

ABSTRACT

The signals that regulate peripheral blood vessel formation during development are still under investigation. The hormone leptin promotes blood vessel formation, adipose tissue establishment and expansion, tumor growth, and wound healing, but the underlying mechanisms for these actions are currently unknown. We investigated whether leptin promotes angiogenesis in the developing tail fin using embryonic transgenic xflk-1:GFP Xenopus laevis, which express a green fluorescent protein on vascular endothelial cells to mark blood vessels. We found that leptin protein is expressed in endothelial cells of developing blood vessels and that leptin treatment via injection increased phosphorylated STAT3 signaling, which is indicative of leptin activation of its receptor, in blood vessels of the larval tail fin. Leptin administration via media increased vessel length, branching, and reconnection with the cardinal vein, while decreased leptin signaling via immunoneutralization had an opposing effect on vessel development. We also observed disorganization of major vessels and microvessels of the tail fin and muscle when leptin signaling was decreased. Reduced leptin signaling lowered mRNA expression of cenpk, gpx1, and mmp9, markers for cell proliferation, antioxidation, and extracellular matrix remodeling/cell migration, respectively, in the developing tail, providing insight into three possible mechanisms underlying leptin's promotion of angiogenesis. Together these results illustrate that leptin levels are correlated with embryonic angiogenesis and that leptin coordinates multiple aspects of blood vessel growth and development, showing that leptin is an important morphogen during embryonic development.


Subject(s)
Larva , Leptin , Neovascularization, Physiologic , Signal Transduction , Tail , Xenopus laevis , Animals , Leptin/metabolism , Tail/blood supply , Tail/embryology , Xenopus laevis/embryology , Xenopus laevis/metabolism , Larva/metabolism , Blood Vessels/embryology , Blood Vessels/metabolism , Xenopus Proteins/metabolism , Xenopus Proteins/genetics , Animals, Genetically Modified , STAT3 Transcription Factor/metabolism , Embryo, Nonmammalian/metabolism , Green Fluorescent Proteins/metabolism , Gene Expression Regulation, Developmental
15.
Mol Cells ; 47(6): 100068, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38759887

ABSTRACT

The coordinated movement of germ layer progenitor cells reaches its peak at the dorsal side, where the Bmp signaling gradient is low, and minimum at the ventral side, where the Bmp gradient is high. This dynamic cell movement is regulated by the interplay of various signaling pathways. The noncanonical Wnt signaling cascade serves as a pivotal regulator of convergence and extension cell movement, facilitated by the activation of small GTPases such as Rho, Rab, and Rac. However, the underlying cause of limited cell movement at the ventral side remains elusive. To explore the functional role of a key regulator in constraining gastrulation cell movement at the ventral side, we investigated the Bmp4-direct target gene, sizzled (szl), to assess its potential role in inhibiting noncanonical Wnt signaling. In our current study, we demonstrated that ectopic expression of szl led to gastrulation defects in a dose-dependent manner without altering cell fate specification. Overexpression of szl resulted in decreased elongation of Activin-treated animal cap and Keller explants. Furthermore, our immunoprecipitation assay unveiled the physical interaction of Szl with noncanonical Wnt ligand proteins (Wnt5 and Wnt11). Additionally, the activation of small GTPases involved in Wnt signaling mediation (RhoA and Rac1) was diminished upon szl overexpression. In summary, our findings suggest that Bmp4 signaling negatively modulates cell movement from the ventral side of the embryo by inducing szl expression during early Xenopus gastrulation.


Subject(s)
Bone Morphogenetic Protein 4 , Cell Movement , Gastrulation , Xenopus Proteins , Xenopus laevis , Animals , Bone Morphogenetic Protein 4/metabolism , Ligands , Wnt Proteins/metabolism , Wnt Signaling Pathway , Xenopus laevis/embryology , Xenopus laevis/metabolism , Xenopus Proteins/metabolism , Xenopus Proteins/genetics
16.
Cells Dev ; 179: 203918, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38574816

ABSTRACT

Increased brain size and its rostral bias are hallmarks of vertebrate evolution, but the underlying developmental and genetic basis remains poorly understood. To provide clues to understanding vertebrate brain evolution, we investigated the developmental mechanisms of brain enlargement observed in the offspring of a previously unrecognized, spontaneously occurring female variant line of Xenopus that appears to reflect a genetic variation. Brain enlargement in larvae from this line showed a pronounced rostral bias that could be traced back to the neural plate, the primordium of the brain. At the gastrula stage, the Spemann organizer, which is known to induce the neural plate from the adjacent dorsal ectoderm and give it the initial rostrocaudal patterning, was expanded from dorsal to ventral in a large proportion of the offspring of variant females. Consistently, siamois expression, which is required for Spemann organizer formation, was expanded laterally from dorsal to ventral at the blastula stage in variant offspring. This implies that the active region of the Wnt/ß-catenin signaling pathway was similarly expanded in advance on the dorsal side, as siamois is a target gene of this pathway. Notably, the earliest detectable change in variant offspring was in fertilized eggs, in which maternal wnt11b mRNA, a candidate dorsalizing factor responsible for activating Wnt/ß-catenin signaling in the dorsal embryonic region, had a wider distribution in the vegetal cortical cytoplasm. Since lateral spreading of wnt11b mRNA, and possibly that of other potential maternal dorsalizing factors in these eggs, is expected to facilitate lateral expansion of the active region of the Wnt/ß-catenin pathway during subsequent embryonic stages, we concluded that aberrant Wnt/ß-catenin signaling could cause rostral-biased brain enlargement via expansion of siamois expression and consequent expansion of the Spemann organizer in Xenopus. Our studies of spontaneously occurring variations in brain development in Xenopus would provide hints for uncovering genetic mutations that drive analogous morphogenetic variations during vertebrate brain evolution.


Subject(s)
Brain , Larva , Wnt Proteins , Wnt Signaling Pathway , Xenopus Proteins , Animals , Female , Wnt Signaling Pathway/genetics , Brain/metabolism , Brain/embryology , Larva/metabolism , Larva/genetics , Xenopus Proteins/metabolism , Xenopus Proteins/genetics , Wnt Proteins/metabolism , Wnt Proteins/genetics , beta Catenin/metabolism , beta Catenin/genetics , Gene Expression Regulation, Developmental , Xenopus laevis/embryology , Xenopus laevis/genetics , Body Patterning/genetics , Embryo, Nonmammalian/metabolism , Organ Size
17.
Dev Biol ; 511: 63-75, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38621649

ABSTRACT

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


Subject(s)
Dyrk Kinases , Xenopus Proteins , Xenopus laevis , Animals , Branchial Region/embryology , Branchial Region/metabolism , Craniofacial Abnormalities/genetics , Craniofacial Abnormalities/embryology , Craniofacial Abnormalities/metabolism , Embryo, Nonmammalian/metabolism , Embryo, Nonmammalian/embryology , Gene Expression Regulation, Developmental , Neural Crest/embryology , Neural Crest/metabolism , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/genetics , Protein-Tyrosine Kinases/metabolism , Protein-Tyrosine Kinases/genetics , Signal Transduction , Xenopus laevis/embryology , Xenopus laevis/metabolism , Xenopus Proteins/metabolism , Xenopus Proteins/genetics
18.
Proc Natl Acad Sci U S A ; 121(19): e2311685121, 2024 May 07.
Article in English | MEDLINE | ID: mdl-38683994

ABSTRACT

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


Subject(s)
Neural Crest , Single-Cell Analysis , Xenopus laevis , Animals , Neural Crest/cytology , Neural Crest/metabolism , Single-Cell Analysis/methods , Xenopus laevis/embryology , Gene Expression Regulation, Developmental , Cell Movement , Gene Regulatory Networks , Transcriptome , Gastrulation , Neural Plate/metabolism , Neural Plate/embryology , Neural Plate/cytology , Epithelial-Mesenchymal Transition/genetics , Embryo, Nonmammalian/metabolism , Embryo, Nonmammalian/cytology , Neurulation/genetics , Neurulation/physiology , Cell Differentiation
19.
J Appl Toxicol ; 44(7): 1040-1049, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38531109

ABSTRACT

As part of the US Environmental Protection Agency's perfluoroalkyl and polyfluoroalkyl substances (PFAS) Action Plan, the agency is committed to increasing our understanding of the potential ecological effects of PFAS. The objective of these studies was to examine the developmental toxicity of PFAS using the laboratory model amphibian species Xenopus laevis. We had two primary aims: (1) to understand the developmental toxicity of a structurally diverse set of PFAS compounds in developing embryos and (2) to characterize the potential impacts of perfluorooctanesulfonic acid (PFOS), perfluorohexanesulfonic acid (PFHxS), perfluorooctanoic acid (PFOA), and hexafluoropropylene oxide-dimer acid (HFPO-DA a.k.a. GenX), on growth and thyroid hormone-controlled metamorphosis. We employed a combination of static renewal and flow-through exposure designs. Embryos were exposed to 17 structurally diverse PFAS starting at the midblastula stage through the completion of organogenesis (96 h). To investigate impacts on PFOS, PFOA, PFHxS, and HFPO-DA on development and metamorphosis, larvae were exposed from premetamorphosis (Nieuwkoop Faber stage 51 or 54) through pro metamorphosis. Of the PFAS tested in embryos, only 1H,1H,10H,10H-perfluorodecane-1,10-diol (FC10-diol) and perfluorohexanesulfonamide (FHxSA) exposure resulted in clear concentration-dependent developmental toxicity. For both of these PFAS, a significant increase in mortality was observed at 2.5 and 5 mg/L. For FC10-diol, 100% of the surviving embryos were malformed at 1.25 and 2.5 mg/L, while for FHxSA, a significant increase in malformations (100%) was observed at 2.5 and 5 mg/L. Developmental stage achieved was the most sensitive endpoint with significant effects observed at 1.25 and 0.625 mg/L for FC10-diol and FHxSA, respectively. In larval studies, we observed impacts on growth following exposure to PFHxS and PFOS at concentrations of 100 and 2.5 mg/L, respectively, while no impacts were observed in larvae when exposed to PFOA and HFPO-DA at concentration of 100 mg/L. Further, we did not observe impacts on thyroid endpoints in exposed larvae. These experiments have broadened our understanding of the impact of PFAS on anuran development.


Subject(s)
Embryo, Nonmammalian , Fluorocarbons , Larva , Metamorphosis, Biological , Xenopus laevis , Animals , Xenopus laevis/embryology , Larva/drug effects , Larva/growth & development , Fluorocarbons/toxicity , Embryo, Nonmammalian/drug effects , Metamorphosis, Biological/drug effects , Alkanesulfonic Acids/toxicity , Sulfonic Acids/toxicity , Dose-Response Relationship, Drug , Caprylates/toxicity , Embryonic Development/drug effects , Thyroid Hormones
20.
Dis Model Mech ; 17(6)2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38501224

ABSTRACT

De novo truncating variants in fibrosin-like 1 (FBRSL1), a member of the AUTS2 gene family, cause a disability syndrome, including organ malformations such as heart defects. Here, we use Xenopus laevis to investigate whether Fbrsl1 plays a role in heart development. Xenopus laevis fbrsl1 is expressed in tissues relevant for heart development, and morpholino-mediated knockdown of Fbrsl1 results in severely hypoplastic hearts. Our data suggest that Fbrsl1 is required for the development of the first heart field, which contributes to the ventricle and the atria, but not for the second heart field, which gives rise to the outflow tract. The morphant heart phenotype could be rescued using a human N-terminal FBRSL1 isoform that contains an alternative exon, but lacks the AUTS2 domain. N-terminal isoforms carrying patient variants failed to rescue. Interestingly, a long human FBRSL1 isoform, harboring the AUTS2 domain, also did not rescue the morphant heart defects. Thus, our data suggest that different FBRSL1 isoforms may have distinct functions and that only the short N-terminal isoform, appears to be critical for heart development.


Subject(s)
Heart Defects, Congenital , Heart , Xenopus Proteins , Xenopus laevis , Animals , Humans , Gene Expression Regulation, Developmental , Gene Knockdown Techniques , Heart/embryology , Heart Defects, Congenital/genetics , Heart Defects, Congenital/pathology , Phenotype , Protein Isoforms/metabolism , Protein Isoforms/genetics , Xenopus laevis/embryology , Xenopus Proteins/metabolism , Xenopus Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL