Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 12.790
Filter
1.
Sci Rep ; 14(1): 18212, 2024 08 06.
Article in English | MEDLINE | ID: mdl-39107332

ABSTRACT

Mammalian dentition exhibits distinct heterodonty, with more simple teeth located in the anterior area of the jaw and more complex teeth situated posteriorly. While some region-specific differences in signalling have been described previously, here we performed a comprehensive analysis of gene expression at the early stages of odontogenesis to obtain complete knowledge of the signalling pathways involved in early jaw patterning. Gene expression was analysed separately on anterior and posterior areas of the lower jaw at two early stages (E11.5 and E12.5) of odontogenesis. Gene expression profiling revealed distinct region-specific expression patterns in mouse mandibles, including several known BMP and FGF signalling members and we also identified several new molecules exhibiting significant differences in expression along the anterior-posterior axis, which potentially can play the role during incisor and molar specification. Next, we followed one of the anterior molecules, SATB2, which was expressed not only in the anterior mesenchyme where incisor germs are initiated, however, we uncovered a distinct SATB2-positive region in the mesenchyme closely surrounding molars. Satb2-deficient animals demonstrated defective incisor development confirming a crucial role of SATB2 in formation of anterior teeth. On the other hand, ectopic tooth germs were observed in the molar area indicating differential effect of Satb2-deficiency in individual jaw regions. In conclusion, our data provide a rich source of fundamental information, which can be used to determine molecular regulation driving early embryonic jaw patterning and serve for a deeper understanding of molecular signalling directed towards incisor and molar development.


Subject(s)
Gene Expression Profiling , Gene Expression Regulation, Developmental , Mandible , Matrix Attachment Region Binding Proteins , Odontogenesis , Transcription Factors , Animals , Matrix Attachment Region Binding Proteins/genetics , Matrix Attachment Region Binding Proteins/metabolism , Mice , Mandible/metabolism , Mandible/embryology , Odontogenesis/genetics , Transcription Factors/genetics , Transcription Factors/metabolism , Tooth/metabolism , Tooth/embryology , Tooth/growth & development , Incisor/metabolism , Incisor/embryology , Incisor/growth & development , Body Patterning/genetics , Signal Transduction
2.
Open Biol ; 14(8): 240115, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39165121

ABSTRACT

Crinoids belong to the Echinodermata, marine invertebrates with a highly derived adult pentaradial body plan. As the sister group to all other extant echinoderms, crinoids occupy a key phylogenetic position to explore the evolutionary history of the whole phylum. However, their development remains understudied compared with that of other echinoderms. Therefore, the aim here was to establish the Mediterranean feather star (Antedon mediterranea) as an experimental system for developmental biology. We first set up a method for culturing embryos in vitro and defined a standardized staging system for this species. We then optimized protocols to characterize the morphological and molecular development of the main structures of the feather star body plan. Focusing on the nervous system, we showed that the larval apical organ includes serotonergic, GABAergic and glutamatergic neurons, which develop within a conserved anterior molecular signature. We described the composition of the early post-metamorphic nervous system and revealed that it has an anterior signature. These results further our knowledge on crinoid development and provide new techniques to investigate feather star embryogenesis. This will pave the way for the inclusion of crinoids in comparative studies addressing the origin of the echinoderm body plan and the evolutionary diversification of deuterostomes.


Subject(s)
Echinodermata , Embryonic Development , Nervous System , Animals , Echinodermata/genetics , Echinodermata/embryology , Echinodermata/growth & development , Nervous System/embryology , Nervous System/metabolism , Gene Expression Regulation, Developmental , Embryo, Nonmammalian/metabolism , Phylogeny , Biological Evolution , Larva/growth & development , Body Patterning
3.
Cell Stem Cell ; 31(8): 1097-1098, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-39094539

ABSTRACT

Morphogen gradients are critical regulators of embryonic development. In this issue, Liu et al.1 introduce a microfluidic system that externally applies morphogen gradients to an in vitro model of human embryo segmentation. It enables the investigation of signaling gradients during this developmental process at unprecedented levels of precision.


Subject(s)
Embryonic Development , Humans , Embryo, Mammalian/cytology , Body Patterning , Microfluidics , Animals
4.
Nat Commun ; 15(1): 6547, 2024 Aug 02.
Article in English | MEDLINE | ID: mdl-39095343

ABSTRACT

Myosin1D (Myo1D) has recently emerged as a conserved regulator of animal Left-Right (LR) asymmetry that governs the morphogenesis of the vertebrate central LR Organizer (LRO). In addition to Myo1D, the zebrafish genome encodes the closely related Myo1G. Here we show that while Myo1G also controls LR asymmetry, it does so through an entirely different mechanism. Myo1G promotes the Nodal-mediated transfer of laterality information from the LRO to target tissues. At the cellular level, Myo1G is associated with endosomes positive for the TGFß signaling adapter SARA. myo1g mutants have fewer SARA-positive Activin receptor endosomes and a reduced responsiveness to Nodal ligands that results in a delay of left-sided Nodal propagation and tissue-specific laterality defects in organs that are most distant from the LRO. Additionally, Myo1G promotes signaling by different Nodal ligands in specific biological contexts. Our findings therefore identify Myo1G as a context-dependent regulator of the Nodal signaling pathway.


Subject(s)
Body Patterning , Signal Transduction , Zebrafish Proteins , Zebrafish , Animals , Zebrafish/genetics , Zebrafish/metabolism , Zebrafish/embryology , Zebrafish Proteins/metabolism , Zebrafish Proteins/genetics , Body Patterning/genetics , Nodal Protein/metabolism , Nodal Protein/genetics , Gene Expression Regulation, Developmental , Endosomes/metabolism , Myosins/metabolism , Myosins/genetics , Mutation , Myosin Type I/metabolism , Myosin Type I/genetics , Embryo, Nonmammalian/metabolism
5.
PLoS One ; 19(7): e0293852, 2024.
Article in English | MEDLINE | ID: mdl-39083515

ABSTRACT

The zinc finger and BTB domain-containing 11 gene (zbtb11) is expressed in the Xenopus anterior neuroectoderm, but the molecular nature of the Zbtb11 protein during embryonic development remains to be elucidated. Here, we show the role of Zbtb11 in anterior patterning of the neuroectoderm and the cooperative action with the transcription factor Otx2. Both overexpression and knockdown of zbtb11 caused similar phenotypes: expanded expression of the posterior gene gbx2 in the neural plate, and later microcephaly with reduced eyes, suggesting that a proper level of zbtb11 expression is necessary for normal patterning of the neuroectoderm, including eye formation. Co-immunoprecipitation assays showed that Zbtb11 formed a complex with itself and with a phosphomimetic and repressive form of Otx2, suggesting that Zbtb11 forms a dimer or oligomer and interacts with Otx2 in a phosphorylation-dependent manner. Reporter analysis further showed that Zbtb11 enhanced the activity of the phosphomimetic Otx2 to repress a silencer element of the posterior gene meis3. These data suggest that Zbtb11 coordinates with phosphorylated Otx2 to specify the anterior neuroectoderm by repressing posterior genes.


Subject(s)
Gene Expression Regulation, Developmental , Neural Plate , Otx Transcription Factors , Xenopus Proteins , Animals , Otx Transcription Factors/metabolism , Otx Transcription Factors/genetics , Xenopus Proteins/genetics , Xenopus Proteins/metabolism , Neural Plate/metabolism , Neural Plate/embryology , Xenopus laevis , Protein Binding , Phosphorylation , Body Patterning/genetics
6.
New Phytol ; 243(5): 1887-1898, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38984686

ABSTRACT

The role of maternal tissue in embryogenesis remains enigmatic in many complex organisms. Here, we investigate the contribution of maternal tissue to apical-basal patterning in the kelp embryo. Focussing on Undaria pinnatifida, we studied the effects of detachment from the maternal tissue using microsurgery, staining of cell wall modifications, morphometric measurements, flow cytometry, genotyping and a modified kelp fertilisation protocol synchronising kelp embryogenesis. Detached embryos are rounder and often show aberrant morphologies. When a part of the oogonial cell wall remains attached to the zygote, the apical-basal patterning is rescued. Furthermore, the absence of contact with maternal tissue increases parthenogenesis, highlighting the critical role of maternal signals in the initial stages of development. These results show a key role for the connection to the maternal oogonial cell wall in apical-basal patterning in kelps. This observation is reminiscent of another brown alga, Fucus, where the cell wall directs the cell fate. Our findings suggest a conserved mechanism across phylogenetically distant oogamous lineages, where localised secretion of sulphated F2 fucans mediates the establishment of the apical-basal polarity. In this model, the maternal oogonial cell wall mediates basal cell fate determination by providing an extrinsic patterning cue to the future kelp embryo.


Subject(s)
Cell Wall , Undaria , Undaria/physiology , Cell Wall/metabolism , Body Patterning , Kelp/physiology , Parthenogenesis , Edible Seaweeds
7.
J Bioinform Comput Biol ; 22(3): 2450011, 2024 Jun.
Article in English | MEDLINE | ID: mdl-39036846

ABSTRACT

Recent computational modeling of early fruit fly (Drosophila) development has characterized the degree to which gene regulation networks can be robust to natural variability. In the first few hours of development, broad spatial gradients of maternally derived transcription factors activate embryonic gap genes. These gap patterns determine the subsequent segmented insect body plan through pair-rule gene expression. Gap genes are expressed with greater spatial precision than the maternal patterns. Computational modeling of the gap-gap regulatory interactions provides a mechanistic understanding for this robustness to maternal variability in wild-type (WT) patterning. A long-standing question in evolutionary biology has been how a system which is robust, such as the developmental program creating any particular species' body plan, is also evolvable, i.e. how can a system evolve or speciate, if the WT form is strongly buffered and protected? In the present work, we use the WT model to explore the breakdown of such Waddington-type 'canalization'. What levels of variability will push the system out of the WT form; are there particular pathways in the gene regulatory mechanism which are more susceptible to losing the WT form; and when robustness is lost, what types of forms are most likely to occur (i.e. what forms lie near the WT)? Manipulating maternal effects in several different pathways, we find a common gap 'peak-to-step' pattern transition in the loss of WT. We discuss these results in terms of the evolvability of insect segmentation, and in terms of experimental perturbations and mutations which could test the model predictions. We conclude by discussing the prospects for using continuum models of pattern dynamics to investigate a wider range of evo-devo problems.


Subject(s)
Gene Regulatory Networks , Animals , Body Patterning/genetics , Gene Expression Regulation, Developmental , Models, Genetic , Drosophila/genetics , Drosophila/embryology , Computer Simulation , Evolution, Molecular , Biological Evolution , Transcription Factors/genetics , Transcription Factors/metabolism
8.
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
9.
Dev Biol ; 515: 67-78, 2024 Nov.
Article in English | MEDLINE | ID: mdl-38968988

ABSTRACT

Sequence-specific transcription factors often function as components of large regulatory complexes. LIM-domain binding protein (LDB) and single-stranded DNA-binding protein (SSDP) function as core scaffolds of transcriptional complexes in animals and plants. Little is known about potential partners and functions for LDB/SSDP complexes in the context of tissue regeneration. In this work, we find that planarian LDB1 and SSDP2 promote tissue regeneration, with a particular function in anterior regeneration and mediolateral polarity reestablishment. We find that LDB1 and SSDP2 interact with one another and with characterized planarian LIM-HD proteins Arrowhead, Islet1, and Lhx1/5-1. We also show that SSDP2 and LDB1 function with islet1 in polarity reestablishment and with lhx1/5-1 in serotonergic neuron maturation. Finally, we find new roles for LDB1 and SSDP2 in regulating gene expression in the planarian intestine and parenchyma; these functions are likely LIM-HD-independent. Together, our work provides insight into LDB/SSDP complexes in a highly regenerative organism. Further, our work provides a strong starting point for identifying and characterizing potential binding partners of LDB1 and SSDP2 and for exploring roles for these proteins in diverse aspects of planarian physiology.


Subject(s)
Body Patterning , Planarians , Regeneration , Transcription Factors , Animals , Planarians/genetics , Planarians/physiology , Regeneration/genetics , Regeneration/physiology , Transcription Factors/metabolism , Transcription Factors/genetics , Body Patterning/genetics , DNA-Binding Proteins/metabolism , DNA-Binding Proteins/genetics , Helminth Proteins/genetics , Helminth Proteins/metabolism , LIM Domain Proteins/metabolism , LIM Domain Proteins/genetics , LIM-Homeodomain Proteins/metabolism , LIM-Homeodomain Proteins/genetics , Gene Expression Regulation, Developmental
10.
Dev Biol ; 515: 121-128, 2024 Nov.
Article in English | MEDLINE | ID: mdl-39029570

ABSTRACT

Regenerating tissues must remember or interpret their spatial position, using this information to restore original size and patterning. The external skeleton of the zebrafish caudal fin is composed of 18 rays; after any portion of the fin is amputated, position-dependent regenerative growth restores each ray to its original length. We tested for transcriptional differences during regeneration of proximal versus distal tissues and identified 489 genes that differed in proximodistal expression. Thyroid hormone directs multiple aspects of ray patterning along the proximodistal axis, and we identified 364 transcripts showing a proximodistal expression pattern that was dependent on thyroid hormone context. To test what aspects of ray positional identity are directed by extrinsic environental cues versus remembered identity autonomous to the tissue, we transplanted distal portions of rays to proximal environments and evaluated regeneration within the new location. Native regenerating proximal tissue showed robust expression of scpp7, a transcript with thyroid-regulated proximal enrichment; in contrast, regenerating rays originating from transplanted distal tissue showed reduced (distal-like) expression during outgrowth. These distal-to-proximal transplants regenerated far beyond the length of the graft itself, indicating that cues from the proximal environment promoted additional growth. Nonetheless, these transplants initiated regeneration at a much slower rate compared to controls, suggesting memory of distal identity was retained by the transplanted tissue. This early growth retardation caused rays that originated from transplants to grow noticeably shorter than neighboring native rays. While several aspects of fin ray morphology (bifurcation, segment length) were found to be determined by the environment, we found that both regeneration speed and ray length are remembered autonomously by tissues, and that persist through multiple rounds of amputation and regeneration.


Subject(s)
Animal Fins , Regeneration , Zebrafish Proteins , Zebrafish , Animals , Animal Fins/physiology , Regeneration/physiology , Zebrafish Proteins/metabolism , Zebrafish Proteins/genetics , Body Patterning/genetics , Body Patterning/physiology , Thyroid Hormones/metabolism , Gene Expression Regulation, Developmental , Signal Transduction/physiology
11.
Dev Biol ; 515: 92-101, 2024 Nov.
Article in English | MEDLINE | ID: mdl-39029571

ABSTRACT

Congenital lung malformations are fatal at birth in their severe forms. Prevention and early intervention of these birth defects require a comprehensive understanding of the molecular mechanisms of lung development. We find that the loss of inturned (Intu), a cilia and planar polarity effector gene, severely disrupts growth and branching morphogenesis of the mouse embryonic lungs. Consistent with our previous results indicating an important role for Intu in ciliogenesis and hedgehog (Hh) signaling, we find greatly reduced number of primary cilia in both the epithelial and mesenchymal tissues of the lungs. We also find significantly reduced expression of Gli1 and Ptch1, direct targets of Hh signaling, suggesting disruption of cilia-dependent Hh signaling in Intu mutant lungs. An agonist of the Hh pathway activator, smoothened, increases Hh target gene expression and tubulogenesis in explanted wild type, but not Intu mutant, lungs, suggesting impaired Hh signaling response underlying lung morphogenetic defects in Intu mutants. Furthermore, removing both Gli2 and Intu completely abolishes branching morphogenesis of the lung, strongly supporting a mechanism by which Intu regulates lung growth and patterning through cilia-dependent Hh signaling. Moreover, a transcriptomics analysis identifies around 200 differentially expressed genes (DEGs) in Intu mutant lungs, including known Hh target genes Gli1, Ptch1/2 and Hhip. Genes involved in muscle differentiation and function are highly enriched among the DEGs, consistent with an important role of Hh signaling in airway smooth muscle differentiation. In addition, we find that the difference in gene expression between the left and right lungs diminishes in Intu mutants, suggesting an important role of Intu in asymmetrical growth and patterning of the mouse lungs.


Subject(s)
Cilia , Gene Expression Regulation, Developmental , Hedgehog Proteins , Lung , Signal Transduction , Animals , Mice , Body Patterning/genetics , Cilia/metabolism , Hedgehog Proteins/metabolism , Hedgehog Proteins/genetics , Lung/embryology , Lung/metabolism , Morphogenesis/genetics , Patched-1 Receptor/metabolism , Patched-1 Receptor/genetics , Zinc Finger Protein GLI1/metabolism , Zinc Finger Protein GLI1/genetics , Zinc Finger Protein Gli2/metabolism , Zinc Finger Protein Gli2/genetics
12.
Dev Biol ; 515: 151-159, 2024 Nov.
Article in English | MEDLINE | ID: mdl-39067503

ABSTRACT

Many salamanders can completely regenerate a fully functional limb. Limb regeneration is a carefully coordinated process involving several defined stages. One key event during the regeneration process is the patterning of the blastema to inform cells of what they must differentiate into. Although it is known that many genes involved in the initial development of the limb are re-used during regeneration, the exact molecular circuitry involved in this process is not fully understood. Several large-scale transcriptional profiling studies of axolotl limb regeneration have identified many transcription factors that are up-regulated after limb amputation. Sall4 is a transcription factor that has been identified to play essential roles in maintaining cells in an undifferentiated state during development and also plays a unique role in limb development. Inactivation of Sall4 during limb bud development results in defects in anterior-posterior patterning of the limb. Sall4 has been found to be up-regulated during limb regeneration in both Xenopus and salamanders, but to date it function has been untested. We confirmed that Sall4 is up-regulated during limb regeneration in the axolotl using qRT-PCR and identified that it is present in the skin cells and also in cells within the blastema. Using CRISPR technology we microinjected gRNAs specific for Sall4 complexed with cas9 protein into the blastema to specifically knockout Sall4 in blastema cells only. This resulted in limb regenerate defects, including missing digits, fusion of digit elements, and defects in the radius and ulna. This suggests that during regeneration Sall4 may play a similar role in regulating the specification of anterior-proximal skeletal elements.


Subject(s)
Ambystoma mexicanum , Body Patterning , Extremities , Regeneration , Transcription Factors , Animals , Regeneration/genetics , Regeneration/physiology , Transcription Factors/metabolism , Transcription Factors/genetics , Extremities/physiology , Extremities/embryology , Ambystoma mexicanum/genetics , Ambystoma mexicanum/physiology , Body Patterning/genetics , Gene Expression Regulation, Developmental/genetics , Amphibian Proteins/genetics , Amphibian Proteins/metabolism
13.
Open Biol ; 14(7): 240139, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38955223

ABSTRACT

The vertebrate organizer plays a crucial role in building the main (antero-posterior) axis of the embryo: it neuralizes the surrounding ectoderm, and is the site of emigration for cells making axial and paraxial mesendoderm during elongation. The chick organizer becomes a stem zone at the onset of elongation; it stops recruiting cells from the neighbouring ectoderm and generates all its derivatives from the small number of resident cells it contains at the end of gastrulation stages. Nothing is known about the molecular identity of this stem zone. Here, we specifically labelled long-term resident cells of the organizer and compared their RNA-seq profile to that of the neighbouring cell populations. Screening by reverse transcription-polymerase chain reaction and in situ hybridization identified four genes (WIF1, PTGDS, ThPO and UCKL1) that are upregulated only in the organizer region when it becomes a stem zone and remain expressed there during axial elongation. In experiments specifically labelling the resident cells of the mature organizer, we show that only these cells express these genes. These findings molecularly define the organizer as a stem zone and offer a key to understanding how this zone is set up, the molecular control of its cells' behaviour and the evolution of axial growth zones.


Subject(s)
Gene Expression Regulation, Developmental , Organizers, Embryonic , Animals , Chick Embryo , Organizers, Embryonic/metabolism , Body Patterning/genetics , Gastrulation/genetics , Transcriptome , Gene Expression Profiling
14.
Development ; 151(14)2024 Jul 15.
Article in English | MEDLINE | ID: mdl-38940461

ABSTRACT

The vertebral column is a characteristic structure of vertebrates. Genetic studies in mice have shown that Hox-mediated patterning plays a key role in specifying discrete anatomical regions of the vertebral column. Expression pattern analyses in several vertebrate embryos have provided correlative evidence that the anterior boundaries of Hox expression coincide with distinct anatomical vertebrae. However, because functional analyses have been limited to mice, it remains unclear which Hox genes actually function in vertebral patterning in other vertebrates. In this study, various zebrafish Hox mutants were generated for loss-of-function phenotypic analysis to functionally decipher the Hox code responsible for the zebrafish anterior vertebrae between the occipital and thoracic vertebrae. We found that Hox genes in HoxB- and HoxC-related clusters participate in regulating the morphology of the zebrafish anterior vertebrae. In addition, medaka hoxc6a was found to be responsible for anterior vertebral identity, as in zebrafish. Based on phenotypic similarities with Hoxc6 knockout mice, our results suggest that the Hox patterning system, including at least Hoxc6, may have been functionally established in the vertebral patterning of the common ancestor of ray-finned and lobe-finned fishes.


Subject(s)
Body Patterning , Gene Expression Regulation, Developmental , Homeodomain Proteins , Spine , Zebrafish Proteins , Zebrafish , Animals , Zebrafish/genetics , Zebrafish/embryology , Spine/embryology , Body Patterning/genetics , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism , Genes, Homeobox/genetics , Oryzias/genetics , Oryzias/embryology , Mice
15.
Development ; 151(14)2024 Jul 15.
Article in English | MEDLINE | ID: mdl-38940473

ABSTRACT

The direction of left-right visceral asymmetry is conserved in vertebrates. Deviations of the standard asymmetric pattern are rare, and the underlying mechanisms are not understood. Here, we use the teleost Astyanax mexicanus, consisting of surface fish with normal left-oriented heart asymmetry and cavefish with high levels of reversed right-oriented heart asymmetry, to explore natural changes in asymmetry determination. We show that Sonic Hedgehog (Shh) signaling is increased at the posterior midline, Kupffer's vesicle (the teleost left-right organizer) is enlarged and contains longer cilia, and the number of dorsal forerunner cells is increased in cavefish. Furthermore, Shh increase in surface fish embryos induces asymmetric changes resembling the cavefish phenotype. Asymmetric expression of the Nodal antagonist Dand5 is equalized or reversed in cavefish, and Shh increase in surface fish mimics changes in cavefish dand5 asymmetry. Shh decrease reduces the level of right-oriented heart asymmetry in cavefish. Thus, naturally occurring modifications in cavefish heart asymmetry are controlled by the effects of Shh signaling on left-right organizer function.


Subject(s)
Body Patterning , Heart , Hedgehog Proteins , Signal Transduction , Animals , Hedgehog Proteins/metabolism , Hedgehog Proteins/genetics , Body Patterning/genetics , Heart/embryology , Characidae/embryology , Characidae/genetics , Gene Expression Regulation, Developmental , Cilia/metabolism , Embryo, Nonmammalian/metabolism
16.
Cell ; 187(13): 3165-3186, 2024 Jun 20.
Article in English | MEDLINE | ID: mdl-38906093

ABSTRACT

Patterned morphologies, such as segments, spirals, stripes, and spots, frequently emerge during embryogenesis through self-organized coordination between cells. Yet, complex patterns also emerge in adults, suggesting that the capacity for spontaneous self-organization is a ubiquitous property of biological tissues. We review current knowledge on the principles and mechanisms of self-organized patterning in embryonic tissues and explore how these principles and mechanisms apply to adult tissues that exhibit features of patterning. We discuss how and why spontaneous pattern generation is integral to homeostasis and healing of tissues, illustrating it with examples from regenerative biology. We examine how aberrant self-organization underlies diverse pathological states, including inflammatory skin disorders and tumors. Lastly, we posit that based on such blueprints, targeted engineering of pattern-driving molecular circuits can be leveraged for synthetic biology and the generation of organoids with intricate patterns.


Subject(s)
Body Patterning , Animals , Humans , Embryonic Development , Homeostasis , Organoids/metabolism , Aging
17.
PLoS Biol ; 22(6): e3002701, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38913712

ABSTRACT

In the highly regulative embryo of the sea urchin Paracentrotus lividus, establishment of the dorsal-ventral (D/V) axis critically depends on the zygotic expression of the TGF-ß nodal in the ventral ectoderm. nodal expression is first induced ubiquitously in the 32-cell embryo and becomes progressively restricted to the presumptive ventral ectoderm by the early blastula stage. This early spatial restriction of nodal expression is independent of Lefty, and instead relies on the activity of Panda, a maternally expressed TGF-ß ligand related to Lefty and Inhibins, which is required maternally for D/V axis specification. However, the mechanism by which Panda restricts the early nodal expression has remained enigmatic and it is not known if Panda works like a BMP ligand by opposing Nodal and antagonizing Smad2/3 signaling, or if it works like Lefty by sequestering an essential component of the Nodal signaling pathway. In this study, we report that Panda functions as an antagonist of the TGF-ß type II receptor ACVRII (Activin receptor type II), which is the only type II receptor for Nodal signaling in the sea urchin and is also a type II receptor for BMP ligands. Inhibiting translation of acvrII mRNA disrupted D/V patterning across all 3 germ layers and caused acvrII morphants to develop with a typical Nodal loss-of-function phenotype. In contrast, embryos overexpressing acvrII displayed strong ectopic Smad1/5/8 signaling at blastula stages and developed as dorsalized larvae, a phenotype very similar to that caused by over activation of BMP signaling. Remarkably, embryos co-injected with acvrII mRNA and panda mRNA did not show ectopic Smad1/5/8 signaling and developed with a largely normal dorsal-ventral polarity. Furthermore, using an axis induction assay, we found that Panda blocks the ability of ACVRII to orient the D/V axis when overexpressed locally. Using co-immunoprecipitation, we showed that Panda physically interacts with ACVRII, as well as with the Nodal co-receptor Cripto, and with TBR3 (Betaglycan), which is a non-signaling receptor for Inhibins in mammals. At the molecular level, we have traced back the antagonistic activity of Panda to the presence of a single proline residue, conserved with all the Lefty factors, in the ACVRII binding motif of Panda, instead of a serine as in most of TGF-ß ligands. Conversion of this proline to a serine converted Panda from an antagonist that opposed Nodal signaling and promoted dorsalization to an agonist that promoted Nodal signaling and triggered ventralization when overexpressed. Finally, using phylogenomics, we analyzed the emergence of the agonist and antagonist form of Panda in the course of evolution. Our data are consistent with the idea that the presence of a serine at that position, like in most TGF-ß, was the ancestral condition and that the initial function of Panda was possibly in promoting and not in antagonizing Nodal signaling. These results highlight the existence of key functional and structural elements conserved between Panda and Lefty, allow to draw an intriguing parallel between sea urchin Panda and mammalian Inhibin α and raise the unexpected possibility that the original function of Panda may have been in activation of the Nodal pathway rather than in its inhibition.


Subject(s)
Activin Receptors, Type II , Body Patterning , Embryo, Nonmammalian , Gene Expression Regulation, Developmental , Nodal Protein , Paracentrotus , Transforming Growth Factor beta , Animals , Transforming Growth Factor beta/metabolism , Body Patterning/genetics , Paracentrotus/embryology , Paracentrotus/metabolism , Paracentrotus/genetics , Activin Receptors, Type II/metabolism , Activin Receptors, Type II/genetics , Nodal Protein/metabolism , Nodal Protein/genetics , Embryo, Nonmammalian/metabolism , Ligands , Signal Transduction
18.
Science ; 384(6700): 1105-1110, 2024 Jun 07.
Article in English | MEDLINE | ID: mdl-38843334

ABSTRACT

Axis formation in fish and amphibians typically begins with a prepattern of maternal gene products. Annual killifish embryogenesis, however, challenges prepatterning models as blastomeres disperse and then aggregate to form the germ layers and body axes. We show that huluwa, a prepatterning factor thought to break symmetry by stabilizing ß-catenin, is truncated and inactive in Nothobranchius furzeri. Nuclear ß-catenin is not selectively stabilized on one side of the blastula but accumulates in cells forming the aggregate. Blocking ß-catenin activity or Nodal signaling disrupts aggregate formation and germ layer specification. Nodal signaling coordinates cell migration, establishing an early role for this signaling pathway. These results reveal a surprising departure from established mechanisms of axis formation: Huluwa-mediated prepatterning is dispensable, and ß-catenin and Nodal regulate morphogenesis.


Subject(s)
Fundulidae , Morphogenesis , Nodal Protein , beta Catenin , Animals , beta Catenin/metabolism , Blastula/metabolism , Body Patterning , Cell Movement , Cell Nucleus/metabolism , Fundulidae/embryology , Fundulidae/metabolism , Germ Layers/metabolism , Nodal Protein/metabolism , Signal Transduction
19.
Dev Biol ; 514: 12-27, 2024 Oct.
Article in English | MEDLINE | ID: mdl-38862087

ABSTRACT

The development of the sea urchin larval body plan is well understood from extensive studies of embryonic patterning. However, fewer studies have investigated the late larval stages during which the unique pentaradial adult body plan develops. Previous work on late larval development highlights major tissue changes leading up to metamorphosis, but the location of specific cell types during juvenile development is less understood. Here, we improve on technical limitations by applying highly sensitive hybridization chain reaction fluorescent in situ hybridization (HCR-FISH) to the fast-developing and transparent sea urchin Lytechinus pictus, with a focus on skeletogenic cells. First, we show that HCR-FISH can be used in L. pictus to precisely localize skeletogenic cells in the rudiment. In doing so, we provide a detailed staging scheme for the appearance of skeletogenic cells around the rudiment prior to and during biomineralization and show that many skeletogenic cells unassociated with larval rods localize outside of the rudiment prior to localizing inside. Second, we show that downstream biomineralization genes have similar expression patterns during larval and juvenile skeletogenesis, suggesting some conservation of skeletogenic mechanisms during development between stages. Third, we find co-expression of blastocoelar and skeletogenic cell markers around juvenile skeleton located outside of the rudiment, which is consistent with data showing that cells from the non-skeletogenic mesoderm embryonic lineage contribute to the juvenile skeletogenic cell lineage. This work sets the foundation for subsequent studies of other cell types in the late larva of L. pictus to better understand juvenile body plan development, patterning, and evolution.


Subject(s)
Larva , Lytechinus , Animals , Lytechinus/embryology , Larva/growth & development , Gene Expression Regulation, Developmental , In Situ Hybridization, Fluorescence , Sea Urchins/embryology , Metamorphosis, Biological , Body Patterning/genetics , Biomineralization
20.
Dev Cell ; 59(12): 1487-1488, 2024 Jun 17.
Article in English | MEDLINE | ID: mdl-38889690

ABSTRACT

In this issue of Developmental Cell, Bolondi et al. systematically assesses neuro-mesodermal progenitor (NMP) dynamics by combining a mouse stem-cell-based embryo model with molecular recording of single cells, shedding light on the dynamics of neural tube and paraxial mesoderm formation during mammalian development.


Subject(s)
Mesoderm , Animals , Mice , Mesoderm/cytology , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Neural Tube/cytology , Neural Tube/embryology , Cell Differentiation/physiology , Stem Cells/cytology , Stem Cells/metabolism , Body Patterning
SELECTION OF CITATIONS
SEARCH DETAIL