Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.884
Filter
1.
Nature ; 586(7827): 151-155, 2020 10.
Article in English | MEDLINE | ID: mdl-32968275

ABSTRACT

CpG methylation by de novo DNA methyltransferases (DNMTs) 3A and 3B is essential for mammalian development and differentiation and is frequently dysregulated in cancer1. These two DNMTs preferentially bind to nucleosomes, yet cannot methylate the DNA wrapped around the nucleosome core2, and they favour the methylation of linker DNA at positioned nucleosomes3,4. Here we present the cryo-electron microscopy structure of a ternary complex of catalytically competent DNMT3A2, the catalytically inactive accessory subunit DNMT3B3 and a nucleosome core particle flanked by linker DNA. The catalytic-like domain of the accessory DNMT3B3 binds to the acidic patch of the nucleosome core, which orients the binding of DNMT3A2 to the linker DNA. The steric constraints of this arrangement suggest that nucleosomal DNA must be moved relative to the nucleosome core for de novo methylation to occur.


Subject(s)
Cryoelectron Microscopy , DNA (Cytosine-5-)-Methyltransferases/chemistry , DNA (Cytosine-5-)-Methyltransferases/metabolism , Nucleosomes/metabolism , Animals , Biocatalysis , Chromatin Assembly and Disassembly , DNA/chemistry , DNA/metabolism , DNA Methylation , DNA Methyltransferase 3A , Histones/chemistry , Histones/genetics , Histones/metabolism , Humans , Models, Molecular , Nucleosomes/chemistry , Protein Binding , Protein Domains , Xenopus/genetics , DNA Methyltransferase 3B
2.
Nature ; 585(7826): 563-568, 2020 09.
Article in English | MEDLINE | ID: mdl-32939088

ABSTRACT

Neural crest cells (NCCs) are migratory, multipotent embryonic cells that are unique to vertebrates and form an array of clade-defining adult features. The evolution of NCCs has been linked to various genomic events, including the evolution of new gene-regulatory networks1,2, the de novo evolution of genes3 and the proliferation of paralogous genes during genome-wide duplication events4. However, conclusive functional evidence linking new and/or duplicated genes to NCC evolution is lacking. Endothelin ligands (Edns) and endothelin receptors (Ednrs) are unique to vertebrates3,5,6, and regulate multiple aspects of NCC development in jawed vertebrates7-10. Here, to test whether the evolution of Edn signalling was a driver of NCC evolution, we used CRISPR-Cas9 mutagenesis11 to disrupt edn, ednr and dlx genes in the sea lamprey, Petromyzon marinus. Lampreys are jawless fishes that last shared a common ancestor with modern jawed vertebrates around 500 million years ago12. Thus, comparisons between lampreys and gnathostomes can identify deeply conserved and evolutionarily flexible features of vertebrate development. Using the frog Xenopus laevis to expand gnathostome phylogenetic representation and facilitate side-by-side analyses, we identify ancient and lineage-specific roles for Edn signalling. These findings suggest that Edn signalling was activated in NCCs before duplication of the vertebrate genome. Then, after one or more genome-wide duplications in the vertebrate stem, paralogous Edn pathways functionally diverged, resulting in NCC subpopulations with different Edn signalling requirements. We posit that this new developmental modularity facilitated the independent evolution of NCC derivatives in stem vertebrates. Consistent with this, differences in Edn pathway targets are associated with differences in the oropharyngeal skeleton and autonomic nervous system of lampreys and modern gnathostomes. In summary, our work provides functional genetic evidence linking the origin and duplication of new vertebrate genes with the stepwise evolution of a defining vertebrate novelty.


Subject(s)
Endothelins/metabolism , Evolution, Molecular , Neural Crest/cytology , Petromyzon/metabolism , Signal Transduction , Xenopus/metabolism , Animals , Bone Development , Bone and Bones/cytology , Bone and Bones/metabolism , Cell Lineage , Endothelins/genetics , Female , Head/growth & development , Heart/growth & development , Larva/growth & development , Ligands , Male , Petromyzon/genetics , Petromyzon/growth & development , Receptors, Endothelin/deficiency , Receptors, Endothelin/genetics , Receptors, Endothelin/metabolism , Xenopus/genetics , Xenopus/growth & development
3.
Nucleic Acids Res ; 52(17): e79, 2024 Sep 23.
Article in English | MEDLINE | ID: mdl-39119924

ABSTRACT

Helitron-like elements (HLEs) are widespread eukaryotic DNA transposons employing a rolling-circle transposition mechanism. Despite their prevalence in fungi, animals, and plant genomes, identifying Helitrons remains a formidable challenge. We introduce HELIANO, a software for annotating and classifying autonomous and non-autonomous HLE sequences from whole genomes. HELIANO overcomes several limitations of existing tools in speed and accuracy, demonstrated through benchmarking and its application to the complex genomes of frogs (Xenopus tropicalis and Xenopus laevis) and rice (Oryza sativa), where it uncovered numerous previously unidentified HLEs. In an extensive analysis of 404 eukaryote genomes, we found HLEs widely distributed across phyla, with exceptions in specific taxa. HELIANO's application led to the discovery of numerous new HLEs in land plants and identified 20 protein domains captured by certain autonomous HLE families. A comprehensive phylogenetic analysis further classified HLEs into two primary clades, HLE1 and HLE2, and revealed nine subgroups, some of which are enriched within specific taxa. The future use of HELIANO promises to improve the global analysis of HLEs across genomes, significantly advancing our understanding of this fascinating transposon superfamily.


Subject(s)
DNA Transposable Elements , Phylogeny , Software , DNA Transposable Elements/genetics , Animals , Oryza/genetics , Oryza/classification , Eukaryota/genetics , Eukaryota/classification , Genome/genetics , Genomics/methods , Xenopus/genetics , Xenopus/classification , Xenopus laevis/genetics , Genome, Plant
4.
Nucleic Acids Res ; 52(10): 5804-5824, 2024 Jun 10.
Article in English | MEDLINE | ID: mdl-38676942

ABSTRACT

MicroRNAs (miRNAs) that share identical or near-identical sequences constitute miRNA families and are predicted to act redundantly. Yet recent evidence suggests that members of the same miRNA family with high sequence similarity might have different roles and that this functional divergence might be rooted in their precursors' sequence. Current knock-down strategies such as antisense oligonucleotides (ASOs) or miRNA sponges cannot distinguish between identical or near identical miRNAs originating from different precursors to allow exploring unique functions of these miRNAs. We here develop a novel strategy based on short 2'-OMe/LNA-modified oligonucleotides to selectively target specific precursor molecules and ablate the production of individual members of miRNA families in vitro and in vivo. Leveraging the highly conserved Xenopus miR-181a family as proof-of-concept, we demonstrate that 2'-OMe/LNA-ASOs targeting the apical region of pre-miRNAs achieve precursor-selective inhibition of mature miRNA-5p production. Furthermore, we extend the applicability of our approach to the human miR-16 family, illustrating its universality in targeting precursors generating identical miRNAs. Overall, our strategy enables efficient manipulation of miRNA expression, offering a powerful tool to dissect the functions of identical or highly similar miRNAs derived from different precursors within miRNA families.


Subject(s)
MicroRNAs , Oligonucleotides , MicroRNAs/genetics , MicroRNAs/metabolism , Animals , Humans , Oligonucleotides/chemistry , Oligonucleotides/genetics , Oligonucleotides, Antisense/chemistry , Oligonucleotides, Antisense/genetics , Oligonucleotides, Antisense/metabolism , RNA Precursors/metabolism , RNA Precursors/genetics , RNA Precursors/chemistry , Xenopus/genetics
5.
Dev Biol ; 511: 76-83, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38614285

ABSTRACT

This paper introduces a single-cell atlas for pivotal developmental stages in Xenopus, encompassing gastrulation, neurulation, and early tailbud. Notably surpassing its predecessors, the new atlas enhances gene mapping, read counts, and gene/cell type nomenclature. Leveraging the latest Xenopus tropicalis genome version, alongside advanced alignment pipelines and machine learning for cell type assignment, this release maintains consistency with previous cell type annotations while rectifying nomenclature issues. Employing an unbiased approach for cell type assignment proves especially apt for embryonic contexts, given the considerable number of non-terminally differentiated cell types. An alternative cell type attribution here adopts a fuzzy, non-deterministic stance, capturing the transient nature of early embryo progenitor cells by presenting an ensemble of types in superposition. The value of the new resource is emphasized through numerous examples, with a focus on previously unexplored germ cell populations where we uncover novel transcription onset features. Offering interactive exploration via a user-friendly web portal and facilitating complete data downloads, this atlas serves as a comprehensive and accessible reference.


Subject(s)
Xenopus , Animals , Xenopus/embryology , Xenopus/genetics , Gastrulation , Embryo, Nonmammalian/cytology , Neurulation/genetics , Neurulation/physiology , Single-Cell Analysis/methods , Gene Expression Regulation, Developmental
6.
EMBO J ; 40(9): e104913, 2021 05 03.
Article in English | MEDLINE | ID: mdl-33555045

ABSTRACT

During vertebrate gastrulation, mesoderm is induced in pluripotent cells, concomitant with dorsal-ventral patterning and establishing of the dorsal axis. We applied single-cell chromatin accessibility and transcriptome analyses to explore the emergence of cellular heterogeneity during gastrulation in Xenopus tropicalis. Transcriptionally inactive lineage-restricted genes exhibit relatively open chromatin in animal caps, whereas chromatin accessibility in dorsal marginal zone cells more closely reflects transcriptional activity. We characterized single-cell trajectories and identified head and trunk organizer cell clusters in early gastrulae. By integrating chromatin accessibility and transcriptome data, we inferred the activity of transcription factors in single-cell clusters and tested the activity of organizer-expressed transcription factors in animal caps, alone or in combination. The expression profile induced by a combination of Foxb1 and Eomes most closely resembles that observed in the head organizer. Genes induced by Eomes, Otx2, or the Irx3-Otx2 combination are enriched for maternally regulated H3K4me3 modifications, whereas Lhx8-induced genes are marked more frequently by zygotically controlled H3K4me3. Taken together, our results show that transcription factors cooperate in a combinatorial fashion in generally open chromatin to orchestrate zygotic gene expression.


Subject(s)
Chromatin/genetics , Single-Cell Analysis/methods , Transcription Factors/metabolism , Xenopus Proteins/genetics , Xenopus/embryology , Animals , Body Patterning , Chromatin/metabolism , Gastrulation , Gene Expression Profiling , Gene Expression Regulation, Developmental , Sequence Analysis, RNA , Xenopus/genetics , Xenopus Proteins/metabolism
7.
Development ; 148(2)2021 01 19.
Article in English | MEDLINE | ID: mdl-33318149

ABSTRACT

Mutations in the RNA helicase DDX3 have emerged as a frequent cause of intellectual disability in humans. Because many individuals carrying DDX3 mutations have additional defects in craniofacial structures and other tissues containing neural crest (NC)-derived cells, we hypothesized that DDX3 is also important for NC development. Using Xenopus tropicalis as a model, we show that DDX3 is required for normal NC induction and craniofacial morphogenesis by regulating AKT kinase activity. Depletion of DDX3 decreases AKT activity and AKT-dependent inhibitory phosphorylation of GSK3ß, leading to reduced levels of ß-catenin and Snai1: two GSK3ß substrates that are crucial for NC induction. DDX3 function in regulating these downstream signaling events during NC induction is likely mediated by RAC1, a small GTPase whose translation depends on the RNA helicase activity of DDX3. These results suggest an evolutionarily conserved role of DDX3 in NC development by promoting AKT activity, and provide a potential mechanism for the NC-related birth defects displayed by individuals harboring mutations in DDX3 and its downstream effectors in this signaling cascade.


Subject(s)
DEAD-box RNA Helicases/metabolism , Neural Crest/embryology , Neural Crest/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Xenopus Proteins/metabolism , Xenopus/embryology , Xenopus/metabolism , Animals , Cartilage/embryology , Cartilage/metabolism , Embryo, Nonmammalian/metabolism , Face/embryology , Gene Expression Regulation, Developmental , Glycogen Synthase Kinase 3 beta/metabolism , Morphogenesis/genetics , Phosphorylation , Protein Stability , Skull/embryology , Skull/metabolism , Snail Family Transcription Factors/metabolism , Wnt Signaling Pathway , Xenopus/genetics , beta Catenin/metabolism , rac1 GTP-Binding Protein/metabolism
8.
Nature ; 553(7688): 337-341, 2018 01 18.
Article in English | MEDLINE | ID: mdl-29320479

ABSTRACT

Hybridization of eggs and sperm from closely related species can give rise to genetic diversity, or can lead to embryo inviability owing to incompatibility. Although central to evolution, the cellular and molecular mechanisms underlying post-zygotic barriers that drive reproductive isolation and speciation remain largely unknown. Species of the African clawed frog Xenopus provide an ideal system to study hybridization and genome evolution. Xenopus laevis is an allotetraploid with 36 chromosomes that arose through interspecific hybridization of diploid progenitors, whereas Xenopus tropicalis is a diploid with 20 chromosomes that diverged from a common ancestor approximately 48 million years ago. Differences in genome size between the two species are accompanied by organism size differences, and size scaling of the egg and subcellular structures such as nuclei and spindles formed in egg extracts. Nevertheless, early development transcriptional programs, gene expression patterns, and protein sequences are generally conserved. Whereas the hybrid produced when X. laevis eggs are fertilized by X. tropicalis sperm is viable, the reverse hybrid dies before gastrulation. Here we apply cell biological tools and high-throughput methods to study the mechanisms underlying hybrid inviability. We reveal that two specific X. laevis chromosomes are incompatible with the X. tropicalis cytoplasm and are mis-segregated during mitosis, leading to unbalanced gene expression at the maternal to zygotic transition, followed by cell-autonomous catastrophic embryo death. These results reveal a cellular mechanism underlying hybrid incompatibility that is driven by genome evolution and contributes to the process by which biological populations become distinct species.


Subject(s)
Chromosomes/genetics , Hybridization, Genetic , Paternal Inheritance/genetics , Xenopus/genetics , Xenopus/metabolism , Animals , Chromosome Segregation , Chromosomes/metabolism , Cytoplasm/genetics , Cytoplasm/metabolism , Embryo Loss/veterinary , Evolution, Molecular , Female , Genetic Speciation , Male , Mitosis , Xenopus laevis/genetics
9.
Nature ; 557(7706): 564-569, 2018 05.
Article in English | MEDLINE | ID: mdl-29769720

ABSTRACT

The four R-spondin secreted ligands (RSPO1-RSPO4) act via their cognate LGR4, LGR5 and LGR6 receptors to amplify WNT signalling1-3. Here we report an allelic series of recessive RSPO2 mutations in humans that cause tetra-amelia syndrome, which is characterized by lung aplasia and a total absence of the four limbs. Functional studies revealed impaired binding to the LGR4/5/6 receptors and the RNF43 and ZNRF3 transmembrane ligases, and reduced WNT potentiation, which correlated with allele severity. Unexpectedly, however, the triple and ubiquitous knockout of Lgr4, Lgr5 and Lgr6 in mice did not recapitulate the known Rspo2 or Rspo3 loss-of-function phenotypes. Moreover, endogenous depletion or addition of exogenous RSPO2 or RSPO3 in triple-knockout Lgr4/5/6 cells could still affect WNT responsiveness. Instead, we found that the concurrent deletion of rnf43 and znrf3 in Xenopus embryos was sufficient to trigger the outgrowth of supernumerary limbs. Our results establish that RSPO2, without the LGR4/5/6 receptors, serves as a direct antagonistic ligand to RNF43 and ZNRF3, which together constitute a master switch that governs limb specification. These findings have direct implications for regenerative medicine and WNT-associated cancers.


Subject(s)
DNA-Binding Proteins/antagonists & inhibitors , Extremities/embryology , Intercellular Signaling Peptides and Proteins/metabolism , Limb Deformities, Congenital/genetics , Receptors, G-Protein-Coupled/metabolism , Ubiquitin-Protein Ligases/antagonists & inhibitors , Animals , DNA-Binding Proteins/metabolism , Female , Fibroblasts , Gene Knockout Techniques , HEK293 Cells , Humans , Intercellular Signaling Peptides and Proteins/genetics , Male , Mice , Oncogene Proteins/antagonists & inhibitors , Oncogene Proteins/metabolism , Phenotype , Receptors, G-Protein-Coupled/deficiency , Ubiquitin-Protein Ligases/metabolism , Xenopus/genetics
10.
Nucleic Acids Res ; 50(D1): D970-D979, 2022 01 07.
Article in English | MEDLINE | ID: mdl-34791383

ABSTRACT

Echinobase (www.echinobase.org) is a third generation web resource supporting genomic research on echinoderms. The new version was built by cloning the mature Xenopus model organism knowledgebase, Xenbase, refactoring data ingestion pipelines and modifying the user interface to adapt to multispecies echinoderm content. This approach leveraged over 15 years of previous database and web application development to generate a new fully featured informatics resource in a single year. In addition to the software stack, Echinobase uses the private cloud and physical hosts that support Xenbase. Echinobase currently supports six echinoderm species, focused on those used for genomics, developmental biology and gene regulatory network analyses. Over 38 000 gene pages, 18 000 publications, new improved genome assemblies, JBrowse genome browser and BLAST + services are available and supported by the development of a new echinoderm anatomical ontology, uniformly applied formal gene nomenclature, and consistent orthology predictions. A novel feature of Echinobase is integrating support for multiple, disparate species. New genomes from the diverse echinoderm phylum will be added and supported as data becomes available. The common code development design of the integrated knowledgebases ensures parallel improvements as each resource evolves. This approach is widely applicable for developing new model organism informatics resources.


Subject(s)
Databases, Genetic , Echinodermata/genetics , Gene Regulatory Networks , Genome , User-Computer Interface , Animals , Echinodermata/classification , Genomics , Internet , Knowledge Bases , Molecular Sequence Annotation , Phylogeny , Xenopus/genetics
11.
Genesis ; 61(5): e23520, 2023 09.
Article in English | MEDLINE | ID: mdl-37318954

ABSTRACT

The endoplasmic reticulum (ER) membrane protein complex (EMC) is essential for the insertion of a wide variety of transmembrane proteins into the plasma membrane across cell types. Each EMC is composed of Emc1-7, Emc10, and either Emc8 or Emc9. Recent human genetics studies have implicated variants in EMC genes as the basis for a group of human congenital diseases. The patient phenotypes are varied but appear to affect a subset of tissues more prominently than others. Namely, craniofacial development seems to be commonly affected. We previously developed an array of assays in Xenopus tropicalis to assess the effects of emc1 depletion on the neural crest, craniofacial cartilage, and neuromuscular function. We sought to extend this approach to additional EMC components identified in patients with congenital malformations. Through this approach, we determine that EMC9 and EMC10 are important for neural crest development and the development of craniofacial structures. The phenotypes observed in patients and our Xenopus model phenotypes similar to EMC1 loss of function likely due to a similar mechanism of dysfunction in transmembrane protein topogenesis.


Subject(s)
Endoplasmic Reticulum , Neural Crest , Animals , Humans , Neural Crest/metabolism , Endoplasmic Reticulum/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Xenopus/genetics , Xenopus/metabolism
12.
Dev Biol ; 483: 157-168, 2022 03.
Article in English | MEDLINE | ID: mdl-35065905

ABSTRACT

Regeneration of complex tissues is initiated by an injury-induced stress response, eventually leading to activation of developmental signaling pathways such as Wnt signaling. How early injury cues are interpreted and coupled to activation of these developmental signals and their targets is not well understood. Here, we show that Hif1α, a stress induced transcription factor, is required for tail regeneration in Xenopus tropicalis. We find that Hif1α is required for regeneration of differentiated axial tissues, including axons and muscle. Using RNA-sequencing, we find that Hif1α and Wnt converge on a broad set of genes required for posterior specification and differentiation, including the posterior hox genes. We further show that Hif1α is required for transcription via a Wnt-responsive element, a function that is conserved in both regeneration and early neural patterning. Our findings indicate that Hif1α has regulatory roles in Wnt target gene expression across multiple tissue contexts.


Subject(s)
Body Patterning/genetics , Gene Expression , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Tail/metabolism , Wnt Proteins/genetics , Wnt Signaling Pathway/genetics , Xenopus Proteins/genetics , Xenopus/genetics , Animals , Axons/metabolism , Gene Expression Regulation, Developmental , Genes, Homeobox , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Larva/genetics , Muscles/metabolism , Regeneration/genetics , Wnt Proteins/metabolism , Xenopus/metabolism , Xenopus Proteins/metabolism
13.
Development ; 147(21)2020 06 22.
Article in English | MEDLINE | ID: mdl-32467234

ABSTRACT

DYRK1A [dual specificity tyrosine-(Y)-phosphorylation-regulated kinase 1 A] is a high-confidence autism risk gene that encodes a conserved kinase. In addition to autism, individuals with putative loss-of-function variants in DYRK1A exhibit microcephaly, intellectual disability, developmental delay and/or congenital anomalies of the kidney and urinary tract. DYRK1A is also located within the critical region for Down syndrome; therefore, understanding the role of DYRK1A in brain development is crucial for understanding the pathobiology of multiple developmental disorders. To characterize the function of this gene, we used the diploid frog Xenopus tropicalis We discover that Dyrk1a is expressed in ciliated tissues, localizes to ciliary axonemes and basal bodies, and is required for ciliogenesis. We also demonstrate that Dyrk1a localizes to mitotic spindles and that its inhibition leads to decreased forebrain size, abnormal cell cycle progression and cell death during brain development. These findings provide hypotheses about potential mechanisms of pathobiology and underscore the utility of X. tropicalis as a model system for understanding neurodevelopmental disorders.


Subject(s)
Brain/anatomy & histology , Cilia/metabolism , Embryo, Nonmammalian/anatomy & histology , Neurodevelopmental Disorders/genetics , Organogenesis/genetics , Protein Serine-Threonine Kinases/genetics , Protein-Tyrosine Kinases/genetics , Xenopus Proteins/genetics , Xenopus/embryology , Xenopus/genetics , Animals , Brain/embryology , Cell Cycle/genetics , Cell Survival , Gene Expression Regulation, Developmental , Genetic Predisposition to Disease , Organ Size , Phenotype , Protein Serine-Threonine Kinases/metabolism , Protein-Tyrosine Kinases/metabolism , Risk Factors , Spindle Apparatus/metabolism , Telencephalon/anatomy & histology , Xenopus Proteins/metabolism
14.
Dev Growth Differ ; 65(9): 591-598, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37750430

ABSTRACT

Amphibians generally have three types of pigment cells, namely, melanophores (black and brown), xanthophores (yellow and red), and iridophores (iridescent). Single knockout of the tyr, slc2a7, and hps6 genes in Xenopus tropicalis results in the absence of melanophores, xanthophores, and iridophores, respectively. The generation of triple- knockout (3KO) X. tropicalis for these three genes could allow for observation of internal organs without sacrificing the animals, which would be transparent due to the absence of pigments. In this study, we generated 3KO X. tropicalis, which is one of the most widely used model amphibians, through crossing of a slc2a7 single-knockout frog with a tyr and hps6 double-knockout frog, followed by intercrossing of their offspring. The 3KO tadpoles had transparent bodies like the nop mutant and the frogs had translucent bodies. This translucency allowed us to observe the heart, lungs, stomach, liver, and digestive tract through the ventral body skin without surgery. After intravital staining, 3KO X. tropicalis showed much clearer fluorescent signals of mineralized tissues compared with the wild type. These 3KO X. tropicalis provide a useful mutant line for continuous observation of internal organs and fluorescent signals in the body. In particular, such 3KO frogs would revolutionize fluorescence monitoring in transgenic tadpoles and frogs expressing fluorescent proteins.


Subject(s)
Melanophores , Pigmentation , Animals , Xenopus/genetics , Xenopus laevis , Pigmentation/genetics , Skin , Anura
15.
Dev Growth Differ ; 65(8): 481-497, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37505799

ABSTRACT

Since CRISPR-based genome editing technology works effectively in the diploid frog Xenopus tropicalis, a growing number of studies have successfully modeled human genetic diseases in this species. However, most of their targets were limited to non-syndromic diseases that exhibit abnormalities in a small fraction of tissues or organs in the body. This is likely because of the complexity of interpreting the phenotypic variations resulting from somatic mosaic mutations generated in the founder animals (crispants). In this study, we attempted to model the syndromic disease campomelic dysplasia (CD) by generating sox9 crispants in X. tropicalis. The resulting crispants failed to form neural crest cells at neurula stages and exhibited various combinations of jaw, gill, ear, heart, and gut defects at tadpole stages, recapitulating part of the syndromic phenotype of CD patients. Genotyping of the crispants with a variety of allelic series of mutations suggested that the heart and gut defects depend primarily on frame-shift mutations expected to be null, whereas the jaw, gill, and ear defects could be induced not only by such mutations but also by in-frame deletion mutations expected to delete part of the jawed vertebrate-specific domain from the encoded Sox9 protein. These results demonstrate that Xenopus crispants are useful for investigating the phenotype-genotype relationships behind syndromic diseases and examining the tissue-specific role of each functional domain within a single protein, providing novel insights into vertebrate jaw evolution.


Subject(s)
Campomelic Dysplasia , Animals , Humans , Xenopus laevis/metabolism , Campomelic Dysplasia/genetics , Xenopus/genetics , Xenopus/metabolism , SOX9 Transcription Factor/genetics , SOX9 Transcription Factor/metabolism , Phenotype , Genotype
16.
Dev Growth Differ ; 65(4): 203-214, 2023 May.
Article in English | MEDLINE | ID: mdl-37127930

ABSTRACT

Cryptochromes (CRYs) are multifunctional molecules that act as a circadian clock oscillating factor, a blue-light sensor, and a light-driven magnetoreceptor. Cry genes are classified into several groups based on the evolutionary relationships. Cryptochrome 6 gene (Cry6) is present in invertebrates and lower vertebrates such as amphibians and fishes. Here we identified a Cry6 ortholog in Xenopus tropicalis (XtCry6). XtCRY6 retains a conserved long N-terminal extension (termed CRY N-terminal extension; CNE) that is not found in any CRY in the other groups. A structural prediction suggested that CNE contained unique structures; a tetrahelical fold structure topologically related to KaiA/RbsU domain, overlapping nuclear- and nucleolar-localizing signals (NLS/NoLS), and a novel motif (termed DI-UIM) overlapping a double-sided ubiquitin-interacting motif (DUIM) and an inverted ubiquitin-interacting motif (IUIM). Potential activities of the NLS/NoLS and DI-UIM were examined to infer the molecular function of XtCRY6. GFP-NLS/NoLS fusion protein exogenously expressed in HEK293 cells was mostly observed in the nucleolus, while GFP-XtCRY6 was observed in the cytoplasm. A glutathione S-transferase (GST) pull-down assay suggested that the DI-UIM physically interacts with polyubiquitin. Consistently, protein docking simulations implied that XtCRY6 DI-UIM binds two ubiquitin molecules in a relationship of a twofold rotational symmetry with the symmetry axis parallel or perpendicular to the DI-UIM helix. These results strongly suggested that XtCRY6 does not function as a circadian transcriptional repressor and that it might have another function such as photoreceptive molecule regulating light-dependent protein degradation or gene expression through a CNE-mediated interaction with ubiquitinated proteins in the cytoplasm and/or nucleolus.


Subject(s)
Cryptochromes , Ubiquitin , Animals , Humans , Cryptochromes/genetics , Cryptochromes/metabolism , Ubiquitin/genetics , Ubiquitin/metabolism , Xenopus/genetics , Xenopus/metabolism , HEK293 Cells , Transcription Factors
17.
Dev Growth Differ ; 65(1): 23-28, 2023 Jan.
Article in English | MEDLINE | ID: mdl-36397722

ABSTRACT

Thyroid hormone (T3) is essential for normal development and metabolism, especially during postembryonic development, a period around birth in mammals when plasma T3 levels reach their peak. T3 functions through two T3 receptors, TRα and TRß. However, little is known about the tissue-specific functions of TRs during postembryonic development because of maternal influence and difficulty in manipulation of mammalian models. We have studied Xenopus tropicalis metamorphosis as a model for human postembryonic development. By using TRα knockout (Xtr·thratmshi ) tadpoles, we have previously shown that TRα is important for T3-dependent intestinal remodeling and hindlimb development but not tail resorption during metamorphosis. Here, we have identified genes bound by TR in premetamorphic wild-type and Xtr·thratmshi tails with or without T3 treatment by using chromatin immunoprecipitation-sequencing and compared them with those in the intestine and hindlimb. Compared to other organs, the tail has much fewer genes bound by TR or affected by TRα knockout. Bioinformatic analyses revealed that among the genes bound by TR in wild-type but not Xtr·thratmshi organs, fewer gene ontology (GO) terms or biological pathways related to metamorphosis were enriched in the tail compared to those in the intestine and hindlimb. This difference likely underlies the drastic effects of TRα knockout on the metamorphosis of the intestine and hindlimb but not the tail. Thus, TRα has tissue-specific roles in regulating T3-dependent anuran metamorphosis by directly targeting the pathways and GO terms important for metamorphosis.


Subject(s)
Thyroid Hormone Receptors alpha , Xenopus Proteins , Xenopus , Animals , Humans , Gene Expression Regulation, Developmental/genetics , Mammals/metabolism , Metamorphosis, Biological/genetics , Thyroid Hormone Receptors alpha/genetics , Thyroid Hormone Receptors alpha/metabolism , Triiodothyronine/genetics , Triiodothyronine/metabolism , Triiodothyronine/pharmacology , Xenopus/genetics , Xenopus/metabolism , Xenopus Proteins/genetics , Xenopus Proteins/metabolism
18.
Nucleic Acids Res ; 49(D1): D254-D260, 2021 01 08.
Article in English | MEDLINE | ID: mdl-33035346

ABSTRACT

Transfer RNA-derived fragments (tRFs) are a new class of small non-coding RNAs and play important roles in biological and physiological processes. Prediction of tRF target genes and binding sites is crucial in understanding the biological functions of tRFs in the molecular mechanisms of human diseases. We developed a publicly accessible web-based database, tRFtarget (http://trftarget.net), for tRF target prediction. It contains the computationally predicted interactions between tRFs and mRNA transcripts using the two state-of-the-art prediction tools RNAhybrid and IntaRNA, including location of the binding sites on the target, the binding region, and free energy of the binding stability with graphic illustration. tRFtarget covers 936 tRFs and 135 thousand predicted targets in eight species. It allows researchers to search either target genes by tRF IDs or tRFs by gene symbols/transcript names. We also integrated the manually curated experimental evidence of the predicted interactions into the database. Furthermore, we provided a convenient link to the DAVID® web server to perform downstream functional pathway analysis and gene ontology annotation on the predicted target genes. This database provides useful information for the scientific community to experimentally validate tRF target genes and facilitate the investigation of the molecular functions and mechanisms of tRFs.


Subject(s)
Databases, Nucleic Acid , RNA, Messenger/genetics , RNA, Small Untranslated/genetics , RNA, Transfer/genetics , Animals , Base Pairing , Base Sequence , Caenorhabditis elegans/genetics , Caenorhabditis elegans/metabolism , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Gene Ontology , Humans , Mice , Molecular Sequence Annotation , Nucleic Acid Conformation , Nucleic Acid Hybridization , RNA, Messenger/chemistry , RNA, Messenger/metabolism , RNA, Small Untranslated/chemistry , RNA, Small Untranslated/metabolism , RNA, Transfer/chemistry , RNA, Transfer/metabolism , Rhodobacter sphaeroides/genetics , Rhodobacter sphaeroides/metabolism , Schizosaccharomyces/genetics , Schizosaccharomyces/metabolism , Thermodynamics , Xenopus/genetics , Xenopus/metabolism , Zebrafish/genetics , Zebrafish/metabolism
19.
PLoS Genet ; 16(11): e1009121, 2020 11.
Article in English | MEDLINE | ID: mdl-33166278

ABSTRACT

In many species, sexual differentiation is a vital prelude to reproduction, and disruption of this process can have severe fitness effects, including sterility. It is thus interesting that genetic systems governing sexual differentiation vary among-and even within-species. To understand these systems more, we investigated a rare example of a frog with three sex chromosomes: the Western clawed frog, Xenopus tropicalis. We demonstrate that natural populations from the western and eastern edges of Ghana have a young Y chromosome, and that a male-determining factor on this Y chromosome is in a very similar genomic location as a previously known female-determining factor on the W chromosome. Nucleotide polymorphism of expressed transcripts suggests genetic degeneration on the W chromosome, emergence of a new Y chromosome from an ancestral Z chromosome, and natural co-mingling of the W, Z, and Y chromosomes in the same population. Compared to the rest of the genome, a small sex-associated portion of the sex chromosomes has a 50-fold enrichment of transcripts with male-biased expression during early gonadal differentiation. Additionally, X. tropicalis has sex-differences in the rates and genomic locations of recombination events during gametogenesis that are similar to at least two other Xenopus species, which suggests that sex differences in recombination are genus-wide. These findings are consistent with theoretical expectations associated with recombination suppression on sex chromosomes, demonstrate that several characteristics of old and established sex chromosomes (e.g., nucleotide divergence, sex biased expression) can arise well before sex chromosomes become cytogenetically distinguished, and show how these characteristics can have lingering consequences that are carried forward through sex chromosome turnovers.


Subject(s)
Sex Chromosomes/genetics , Sex Determination Processes/genetics , Sex Differentiation/genetics , Xenopus/genetics , Animals , Female , Genetic Fitness , Ghana , Male , Recombination, Genetic
20.
PLoS Genet ; 16(8): e1008967, 2020 08.
Article in English | MEDLINE | ID: mdl-32813698

ABSTRACT

Dysregulation of ribosome production can lead to a number of developmental disorders called ribosomopathies. Despite the ubiquitous requirement for these cellular machines used in protein synthesis, ribosomopathies manifest in a tissue-specific manner, with many affecting the development of the face. Here we reveal yet another connection between craniofacial development and making ribosomes through the protein Paired Box 9 (PAX9). PAX9 functions as an RNA Polymerase II transcription factor to regulate the expression of proteins required for craniofacial and tooth development in humans. We now expand this function of PAX9 by demonstrating that PAX9 acts outside of the cell nucleolus to regulate the levels of proteins critical for building the small subunit of the ribosome. This function of PAX9 is conserved to the organism Xenopus tropicalis, an established model for human ribosomopathies. Depletion of pax9 leads to craniofacial defects due to abnormalities in neural crest development, a result consistent with that found for depletion of other ribosome biogenesis factors. This work highlights an unexpected layer of how the making of ribosomes is regulated in human cells and during embryonic development.


Subject(s)
Developmental Disabilities/genetics , Embryonic Development/genetics , PAX9 Transcription Factor/genetics , Ribosomes/genetics , Animals , Cell Nucleolus/genetics , Developmental Disabilities/pathology , Gene Expression Regulation, Developmental/genetics , Humans , Neural Crest/growth & development , Neural Crest/metabolism , Neural Crest/pathology , Protein Biosynthesis/genetics , RNA Polymerase II/genetics , Ribosomes/pathology , Xenopus/genetics , Xenopus/growth & development
SELECTION OF CITATIONS
SEARCH DETAIL