Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.344
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.
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
3.
J Radiat Res ; 65(3): 315-322, 2024 May 23.
Article in English | MEDLINE | ID: mdl-38648785

ABSTRACT

Ionizing radiation (IR) causes DNA damage, particularly DNA double-strand breaks (DSBs), which have significant implications for genome stability. The major pathways of repairing DSBs are homologous recombination (HR) and nonhomologous end joining (NHEJ). However, the repair mechanism of IR-induced DSBs in embryos is not well understood, despite extensive research in somatic cells. The externally developing aquatic organism, Xenopus tropicalis, serves as a valuable model for studying embryo development. A significant increase in zygotic transcription occurs at the midblastula transition (MBT), resulting in a longer cell cycle and asynchronous cell divisions. This study examines the impact of X-ray irradiation on Xenopus embryos before and after the MBT. The findings reveal a heightened X-ray sensitivity in embryos prior to the MBT, indicating a distinct shift in the DNA repair pathway during embryo development. Importantly, we show a transition in the dominant DSB repair pathway from NHEJ to HR before and after the MBT. These results suggest that the MBT plays a crucial role in altering DSB repair mechanisms, thereby influencing the IR sensitivity of developing embryos.


Subject(s)
Blastula , DNA Breaks, Double-Stranded , DNA Repair , Animals , DNA Breaks, Double-Stranded/radiation effects , DNA Repair/radiation effects , Blastula/radiation effects , Blastula/metabolism , Xenopus/embryology , DNA End-Joining Repair/radiation effects , Embryo, Nonmammalian/radiation effects , Embryo, Nonmammalian/metabolism , X-Rays
4.
Mol Reprod Dev ; 91(8): e23718, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38126950

ABSTRACT

Specification of the germline and its segregation from the soma mark one of the most crucial events in the lifetime of an organism. In different organisms, this specification can occur through either inheritance or inductive mechanisms. In species such as Xenopus and zebrafish, the specification of primordial germ cells relies on the inheritance of maternal germline determinants that are synthesized and sequestered in the germ plasm during oogenesis. In this review, we discuss the formation of the germ plasm, how germline determinants are recruited into the germ plasm during oogenesis, and the dynamics of the germ plasm during oogenesis and early embryonic development.


Subject(s)
Embryonic Development , Oogenesis , Zebrafish , Animals , Oogenesis/physiology , Zebrafish/embryology , Embryonic Development/physiology , Germ Cells/metabolism , Female , Xenopus/embryology
5.
Cell Rep ; 38(7): 110364, 2022 02 15.
Article in English | MEDLINE | ID: mdl-35172134

ABSTRACT

Mesendodermal specification is one of the earliest events in embryogenesis, where cells first acquire distinct identities. Cell differentiation is a highly regulated process that involves the function of numerous transcription factors (TFs) and signaling molecules, which can be described with gene regulatory networks (GRNs). Cell differentiation GRNs are difficult to build because existing mechanistic methods are low throughput, and high-throughput methods tend to be non-mechanistic. Additionally, integrating highly dimensional data composed of more than two data types is challenging. Here, we use linked self-organizing maps to combine chromatin immunoprecipitation sequencing (ChIP-seq)/ATAC-seq with temporal, spatial, and perturbation RNA sequencing (RNA-seq) data from Xenopus tropicalis mesendoderm development to build a high-resolution genome scale mechanistic GRN. We recover both known and previously unsuspected TF-DNA/TF-TF interactions validated through reporter assays. Our analysis provides insights into transcriptional regulation of early cell fate decisions and provides a general approach to building GRNs using highly dimensional multi-omic datasets.


Subject(s)
Endoderm/embryology , Gene Regulatory Networks , Genomics , Mesoderm/embryology , Xenopus/embryology , Xenopus/genetics , Animals , Chromatin/metabolism , Consensus Sequence/genetics , DNA/metabolism , Gastrulation/genetics , Gene Expression Regulation, Developmental , Protein Binding , RNA/metabolism , Transcription Factors/metabolism , Transcription, Genetic
6.
Elife ; 102021 10 13.
Article in English | MEDLINE | ID: mdl-34643182

ABSTRACT

The gene regulatory networks that coordinate the development of the cardiac and pulmonary systems are essential for terrestrial life but poorly understood. The T-box transcription factor Tbx5 is critical for both pulmonary specification and heart development, but how these activities are mechanistically integrated remains unclear. Here using Xenopus and mouse embryos, we establish molecular links between Tbx5 and retinoic acid (RA) signaling in the mesoderm and between RA signaling and sonic hedgehog expression in the endoderm to unveil a conserved RA-Hedgehog-Wnt signaling cascade coordinating cardiopulmonary (CP) development. We demonstrate that Tbx5 directly maintains expression of aldh1a2, the RA-synthesizing enzyme, in the foregut lateral plate mesoderm via an evolutionarily conserved intronic enhancer. Tbx5 promotes posterior second heart field identity in a positive feedback loop with RA, antagonizing a Fgf8-Cyp regulatory module to restrict FGF activity to the anterior. We find that Tbx5/Aldh1a2-dependent RA signaling directly activates shh transcription in the adjacent foregut endoderm through a conserved MACS1 enhancer. Hedgehog signaling coordinates with Tbx5 in the mesoderm to activate expression of wnt2/2b, which induces pulmonary fate in the foregut endoderm. These results provide mechanistic insight into the interrelationship between heart and lung development informing CP evolution and birth defects.


Subject(s)
Aldehyde Dehydrogenase 1 Family/genetics , Gene Expression Regulation, Developmental , Gene Regulatory Networks , Heart/embryology , Lung/embryology , Retinal Dehydrogenase/genetics , T-Box Domain Proteins/genetics , Xenopus Proteins/genetics , Xenopus/embryology , Aldehyde Dehydrogenase 1 Family/metabolism , Animals , Base Sequence , Mesoderm/embryology , Mice , Retinal Dehydrogenase/metabolism , Sequence Alignment , T-Box Domain Proteins/metabolism , Xenopus/genetics , Xenopus/metabolism , Xenopus Proteins/metabolism , Xenopus laevis/genetics , Xenopus laevis/metabolism
7.
Biol Open ; 10(7)2021 07 15.
Article in English | MEDLINE | ID: mdl-34259326

ABSTRACT

Coordinated polarization of cells in the tissue plane, known as planar cell polarity (PCP), is associated with a signaling pathway critical for the control of morphogenetic processes. Although the segregation of PCP components to opposite cell borders is believed to play a critical role in this pathway, whether PCP derives from egg polarity or preexistent long-range gradient, or forms in response to a localized cue, remains a challenging question. Here we investigate the Xenopus neural plate, a tissue that has been previously shown to exhibit PCP. By imaging Vangl2 and Prickle3, we show that PCP is progressively acquired in the neural plate and requires a signal from the posterior region of the embryo. Tissue transplantations indicated that PCP is triggered in the neural plate by a planar cue from the dorsal blastopore lip. The PCP cue did not depend on the orientation of the graft and was distinct from neural inducers. These observations suggest that neuroectodermal PCP is not instructed by a preexisting molecular gradient but induced by a signal from the dorsal blastopore lip.


Subject(s)
Cell Polarity/physiology , Gastrula/embryology , Morphogenesis/physiology , Neural Plate/embryology , Xenopus/embryology , Animals , Signal Transduction
9.
Development ; 148(13)2021 07 01.
Article in English | MEDLINE | ID: mdl-34164654

ABSTRACT

Understanding the mechanisms of embryonic cell cycles is a central goal of developmental biology, as the regulation of the cell cycle must be closely coordinated with other events during early embryogenesis. Quantitative imaging approaches have recently begun to reveal how the cell cycle oscillator is controlled in space and time, and how it is integrated with mechanical signals to drive morphogenesis. Here, we discuss how the Drosophila embryo has served as an excellent model for addressing the molecular and physical mechanisms of embryonic cell cycles, with comparisons to other model systems to highlight conserved and species-specific mechanisms. We describe how the rapid cleavage divisions characteristic of most metazoan embryos require chemical waves and cytoplasmic flows to coordinate morphogenesis across the large expanse of the embryo. We also outline how, in the late cleavage divisions, the cell cycle is inter-regulated with the activation of gene expression to ensure a reliable maternal-to-zygotic transition. Finally, we discuss how precise transcriptional regulation of the timing of mitosis ensures that tissue morphogenesis and cell proliferation are tightly controlled during gastrulation.


Subject(s)
Cell Cycle Checkpoints/physiology , Drosophila/embryology , Embryonic Development/physiology , Animals , CDC2 Protein Kinase , Cell Cycle/genetics , Drosophila Proteins , Embryo, Mammalian , Embryo, Nonmammalian/metabolism , Embryonic Development/genetics , Gene Expression Regulation, Developmental , Mitosis , Morphogenesis , Xenopus/embryology , Zygote/metabolism
10.
Biochem Biophys Res Commun ; 567: 99-105, 2021 08 27.
Article in English | MEDLINE | ID: mdl-34146908

ABSTRACT

During vertebrate development, the formation of the central nervous system (CNS) is initiated by neural induction and patterning of the embryonic ectoderm. We previously reported that Cdc2-like kinase 2 (Clk2) promotes neural development in Xenopus embryos by regulating morphogen signaling. However, the functions of other Clk family members and their roles in early embryonic development remain unknown. Here, we show that in addition to Clk2, Clk1 and Clk3 play a role in the formation of neural tissue in Xenopus. clk1 and clk3 are co-expressed in the developing neural tissue during early Xenopus embryogenesis. We found that overexpression of clk1 and clk3 increases the expression of neural marker genes in ectodermal explants. Furthermore, knockdown experiments showed that clk3 is required for the formation of neural tissues. These results suggest that Xenopus Clk3 plays an essential role in promoting neural development during early embryogenesis.


Subject(s)
Neurogenesis , Protein Serine-Threonine Kinases/genetics , Protein-Tyrosine Kinases/genetics , Xenopus Proteins/genetics , Xenopus/embryology , Animals , Ectoderm/embryology , Ectoderm/metabolism , Embryo, Nonmammalian/embryology , Embryo, Nonmammalian/metabolism , Gene Expression Regulation, Developmental , Xenopus/genetics
11.
Nat Commun ; 12(1): 3358, 2021 06 07.
Article in English | MEDLINE | ID: mdl-34099733

ABSTRACT

Early stages of embryogenesis depend on subcellular localization and transport of maternal mRNA. However, systematic analysis of these processes is hindered by a lack of spatio-temporal information in single-cell RNA sequencing. Here, we combine spatially-resolved transcriptomics and single-cell RNA labeling to perform a spatio-temporal analysis of the transcriptome during early zebrafish development. We measure spatial localization of mRNA molecules within the one-cell stage embryo, which allows us to identify a class of mRNAs that are specifically localized at an extraembryonic position, the vegetal pole. Furthermore, we establish a method for high-throughput single-cell RNA labeling in early zebrafish embryos, which enables us to follow the fate of individual maternal transcripts until gastrulation. This approach reveals that many localized transcripts are specifically transported to the primordial germ cells. Finally, we acquire spatial transcriptomes of two xenopus species and compare evolutionary conservation of localized genes as well as enriched sequence motifs.


Subject(s)
Cell Tracking/methods , Embryo, Nonmammalian/metabolism , RNA, Messenger/genetics , Transcriptome/genetics , Zebrafish/genetics , Animals , Embryo, Nonmammalian/cytology , Embryo, Nonmammalian/embryology , Female , Gene Expression Regulation, Developmental , Oocytes/cytology , Oocytes/metabolism , RNA, Messenger/metabolism , Single-Cell Analysis/methods , Spatio-Temporal Analysis , Species Specificity , Xenopus/embryology , Xenopus/genetics , Xenopus laevis/embryology , Xenopus laevis/genetics , Zebrafish/embryology
12.
Nat Genet ; 53(7): 1075-1087, 2021 07.
Article in English | MEDLINE | ID: mdl-34099928

ABSTRACT

Animal interphase chromosomes are organized into topologically associating domains (TADs). How TADs are formed is not fully understood. Here, we combined high-throughput chromosome conformation capture and gene silencing to obtain insights into TAD dynamics in Xenopus tropicalis embryos. First, TAD establishment in X. tropicalis is similar to that in mice and flies and does not depend on zygotic genome transcriptional activation. This process is followed by further refinements in active and repressive chromatin compartments and the appearance of loops and stripes. Second, within TADs, higher self-interaction frequencies at one end of the boundary are associated with higher DNA occupancy of the architectural proteins CTCF and Rad21. Third, the chromatin remodeling factor ISWI is required for de novo TAD formation. Finally, TAD structures are variable in different tissues. Our work shows that X. tropicalis is a powerful model for chromosome architecture analysis and suggests that chromatin remodeling plays an essential role in de novo TAD establishment.


Subject(s)
Genome , Models, Molecular , Nucleic Acid Conformation , Xenopus/genetics , Animals , Apoptosis Regulatory Proteins/genetics , Cell Cycle Proteins/genetics , Chromatin/genetics , Chromatin/metabolism , Chromatin Assembly and Disassembly , Computational Biology/methods , Embryonic Development/genetics , Gene Expression Regulation, Developmental , Gene Knockdown Techniques , Genomics/methods , Phenotype , Xenopus/embryology , Xenopus Proteins/genetics
13.
Elife ; 102021 06 29.
Article in English | MEDLINE | ID: mdl-34184636

ABSTRACT

How cells count and regulate organelle number is a fundamental question in cell biology. For example, most cells restrict centrioles to two in number and assemble one cilium; however, multiciliated cells (MCCs) synthesize hundreds of centrioles to assemble multiple cilia. Aberration in centriole/cilia number impairs MCC function and can lead to pathological outcomes. Yet how MCCs control centriole number remains unknown. Using Xenopus, we demonstrate that centriole number scales with apical area over a remarkable 40-fold change in size. We find that tensile forces that shape the apical area also trigger centriole amplification based on both cell stretching experiments and disruption of embryonic elongation. Unexpectedly, Piezo1, a mechanosensitive ion channel, localizes near each centriole suggesting a potential role in centriole amplification. Indeed, depletion of Piezo1 affects centriole amplification and disrupts its correlation with the apical area in a tension-dependent manner. Thus, mechanical forces calibrate cilia/centriole number to the MCC apical area via Piezo1. Our results provide new perspectives to study organelle number control essential for optimal cell function.


Subject(s)
Centrioles/physiology , Animals , Biomechanical Phenomena , CRISPR-Cas Systems , Cell Cycle Proteins , Gene Silencing , Ion Channels , Morpholinos , RNA, Messenger , Xenopus/embryology
14.
Elife ; 102021 05 25.
Article in English | MEDLINE | ID: mdl-34032216

ABSTRACT

Morphogenesis is governed by the interplay of molecular signals and mechanical forces across multiple length scales. The last decade has seen tremendous advances in our understanding of the dynamics of protein localization and turnover at subcellular length scales, and at the other end of the spectrum, of mechanics at tissue-level length scales. Integrating the two remains a challenge, however, because we lack a detailed understanding of the subcellular patterns of mechanical properties of cells within tissues. Here, in the context of the elongating body axis of Xenopus embryos, we combine tools from cell biology and physics to demonstrate that individual cell-cell junctions display finely-patterned local mechanical heterogeneity along their length. We show that such local mechanical patterning is essential for the cell movements of convergent extension and is imparted by locally patterned clustering of a classical cadherin. Finally, the patterning of cadherins and thus local mechanics along cell-cell junctions are controlled by Planar Cell Polarity signaling, a key genetic module for CE that is mutated in diverse human birth defects.


Subject(s)
Cadherins/metabolism , Intercellular Junctions/metabolism , Single-Cell Analysis , Xenopus/embryology , Animals , Body Patterning , Cell Polarity , Morphogenesis
15.
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
16.
Nat Commun ; 12(1): 612, 2021 01 27.
Article in English | MEDLINE | ID: mdl-33504787

ABSTRACT

The motile cilia of ependymal cells coordinate their beats to facilitate a forceful and directed flow of cerebrospinal fluid (CSF). Each cilium originates from a basal body with a basal foot protruding from one side. A uniform alignment of these basal feet is crucial for the coordination of ciliary beating. The process by which the basal foot originates from subdistal appendages of the basal body, however, is unresolved. Here, we show FGFR1 Oncogene Partner (FOP) is a useful marker for delineating the transformation of a circular, unpolarized subdistal appendage into a polarized structure with a basal foot. Ankyrin repeat and SAM domain-containing protein 1A (ANKS1A) interacts with FOP to assemble region I of the basal foot. Importantly, disruption of ANKS1A reduces the size of region I. This produces an unstable basal foot, which disrupts rotational polarity and the coordinated beating of cilia in young adult mice. ANKS1A deficiency also leads to severe degeneration of the basal foot in aged mice and the detachment of cilia from their basal bodies. This role of ANKS1A in the polarization of the basal foot is evolutionarily conserved in vertebrates. Thus, ANKS1A regulates FOP to build and maintain the polarity of subdistal appendages.


Subject(s)
Cilia/metabolism , Molecular Dynamics Simulation , Adaptor Proteins, Signal Transducing/deficiency , Adaptor Proteins, Signal Transducing/metabolism , Aging/pathology , Animals , Basal Bodies/metabolism , Biological Evolution , Cilia/ultrastructure , Embryo, Nonmammalian/metabolism , Mice, Inbred C57BL , Mice, Knockout , Protein Binding , Transcription Factors/metabolism , Xenopus/embryology , Xenopus/metabolism
18.
Dev Biol ; 473: 59-70, 2021 05.
Article in English | MEDLINE | ID: mdl-33484704

ABSTRACT

Xenopus tadpoles are a unique model for regeneration in that they exhibit two distinct phases of age-specific regenerative competence. In Xenopus laevis, young tadpoles fully regenerate following major injuries such as tail transection, then transiently lose regenerative competence during the "refractory period" from stages 45-47. Regenerative competence is then regained in older tadpoles before being permanently lost during metamorphosis. Here we show that a similar refractory period exists in X. tropicalis. Notably, tadpoles lose regenerative competence gradually in X. tropicalis, with full regenerative competence lost at stage 47. We find that the refractory period coincides closely with depletion of maternal yolk stores and the onset of independent feeding, and so we hypothesized that it might be caused in part by nutrient stress. In support of this hypothesis, we find that cell proliferation declines throughout the tail as the refractory period approaches. When we block nutrient mobilization by inhibiting mTOR signaling, we find that tadpole growth and regeneration are reduced, while yolk stores persist. Finally, we are able to restore regenerative competence and cell proliferation during the refractory period by abundantly feeding tadpoles. Our study argues that nutrient stress contributes to lack of regenerative competence and introduces the X. tropicalis refractory period as a valuable new model for interrogating how metabolic constraints inform regeneration.


Subject(s)
Regeneration/physiology , Tail/physiology , Xenopus/embryology , Animals , Cell Proliferation , Egg Yolk , Larva/metabolism , Metamorphosis, Biological/physiology , Nutrients , Signal Transduction , Xenopus/metabolism , Xenopus Proteins/metabolism
19.
Thyroid ; 31(1): 128-142, 2021 01.
Article in English | MEDLINE | ID: mdl-32515287

ABSTRACT

Background: There are two highly conserved thyroid hormone (triiodothyronine [T3]) receptor (TR) genes, TRα and TRß, in all vertebrates, and the expression of TRα but not TRß is activated earlier than T3 synthesis during development. In human, high levels of T3 are present during the several months around birth, and T3 deficiency during this period causes severe developmental abnormalities including skeletal and intestinal defects. It is, however, difficult to study this period in mammals as the embryos and neonates depend on maternal supply of nutrients for survival. However, Xenopus tropicalis undergoes a T3-dependent metamorphosis, which drastically changes essentially every organ in a tadpole. Of interest is intestinal remodeling, which involves near complete degeneration of the larval epithelium through apoptosis. Concurrently, adult intestinal stem cells are formed de novo and subsequently give rise to the self-renewing adult epithelial system, resembling intestinal maturation around birth in mammals. We have previously demonstrated that T3 signaling is essential for the formation of adult intestinal stem cells during metamorphosis. Methods: We studied the function of endogenous TRα in the tadpole intestine by using knockout animals and RNA-seq analysis. Results: We observed that removing endogenous TRα caused defects in intestinal remodeling, including drastically reduced larval epithelial cell death and adult intestinal stem cell proliferation. Using RNA-seq on intestinal RNA from premetamorphic wild-type and TRα-knockout tadpoles treated with or without T3 for one day, before any detectable T3-induced cell death and stem cell formation in the tadpole intestine, we identified more than 1500 genes, which were regulated by T3 treatment of the wild-type but not TRα-knockout tadpoles. Gene Ontology and biological pathway analyses revealed that surprisingly, these TRα-regulated genes were highly enriched with cell cycle-related genes, in addition to genes related to stem cells and apoptosis. Conclusions: Our findings suggest that TRα-mediated T3 activation of the cell cycle program is involved in larval epithelial cell death and adult epithelial stem cell development during intestinal remodeling.


Subject(s)
Adult Stem Cells/metabolism , Cell Cycle , Cell Proliferation , Epithelial Cells/metabolism , Intestinal Mucosa/metabolism , Thyroid Hormone Receptors alpha/deficiency , Triiodothyronine/metabolism , Xenopus Proteins/deficiency , Xenopus/metabolism , Adult Stem Cells/pathology , Animals , Apoptosis , Epithelial Cells/pathology , Gene Expression Regulation, Developmental , Intestinal Mucosa/pathology , Larva/genetics , Larva/metabolism , Metamorphosis, Biological , Signal Transduction , Thyroid Hormone Receptors alpha/genetics , Xenopus/embryology , Xenopus/genetics , Xenopus Proteins/genetics
20.
Dev Biol ; 469: 46-53, 2021 01 01.
Article in English | MEDLINE | ID: mdl-33065118

ABSTRACT

Ciliopathies affect a variety of tissues during development including the heart, kidneys, respiratory tract, and retina. Though an increasing number of monogenic causes of ciliopathies have been described, many remain unexplained. Recently, recessive variants in NUP93 and NUP205 encoding two proteins of the inner ring of the nuclear pore complex were implicated as causes of steroid resistant nephrotic syndrome. In addition, we previously found that the inner ring nucleoporins NUP93 and NUP188 function in proper left-right patterning in developing embryos via a role at the cilium. Here, we describe the role of an additional inner ring nucleoporin NUP205 in cilia biology and establishment of normal organ situs. Using knockdown in Xenopus, we show that Nup205 depletion results in loss of cilia and abnormal cardiac morphology. Furthermore, by transmission electron microscopy, we observe a loss of cilia and mispositioning of intracellular ciliary structures such as basal bodies and rootlets upon depleting inner ring nucleoporins. We describe a model wherein NUP93 interacting with either NUP188 or NUP205 is necessary for cilia. We thus provide evidence that dysregulation of inner ring nucleoporin genes that have been identified in patients may contribute to pathogenesis through cilia dysfunction.


Subject(s)
Cilia/physiology , Nuclear Pore Complex Proteins/physiology , Xenopus Proteins/physiology , Animals , Body Patterning , Cilia/ultrastructure , Epidermis/embryology , Epidermis/ultrastructure , Gene Knockdown Techniques , Heart Defects, Congenital/genetics , Humans , Nuclear Pore Complex Proteins/genetics , Pronephros/ultrastructure , Xenopus/embryology , Xenopus Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL