Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 117
Filter
1.
Nucleic Acids Res ; 51(12): 6190-6207, 2023 07 07.
Article in English | MEDLINE | ID: mdl-37178005

ABSTRACT

Heterochromatin is a key architectural feature of eukaryotic chromosomes critical for cell type-specific gene expression and genome stability. In the mammalian nucleus, heterochromatin segregates from transcriptionally active genomic regions and exists in large, condensed, and inactive nuclear compartments. However, the mechanisms underlying the spatial organization of heterochromatin need to be better understood. Histone H3 lysine 9 trimethylation (H3K9me3) and lysine 27 trimethylation (H3K27me3) are two major epigenetic modifications that enrich constitutive and facultative heterochromatin, respectively. Mammals have at least five H3K9 methyltransferases (SUV39H1, SUV39H2, SETDB1, G9a and GLP) and two H3K27 methyltransferases (EZH1 and EZH2). In this study, we addressed the role of H3K9 and H3K27 methylation in heterochromatin organization using a combination of mutant cells for five H3K9 methyltransferases and an EZH1/2 dual inhibitor, DS3201. We showed that H3K27me3, which is normally segregated from H3K9me3, was redistributed to regions targeted by H3K9me3 after the loss of H3K9 methylation and that the loss of both H3K9 and H3K27 methylation resulted in impaired condensation and spatial organization of heterochromatin. Our data demonstrate that the H3K27me3 pathway safeguards heterochromatin organization after the loss of H3K9 methylation in mammalian cells.


Subject(s)
Epigenesis, Genetic , Heterochromatin , Animals , Heterochromatin/genetics , Histones/metabolism , Lysine/metabolism , Mammals/genetics , Methylation , Histone Methyltransferases/metabolism
2.
Evol Dev ; 25(3): 197-208, 2023 05.
Article in English | MEDLINE | ID: mdl-36946416

ABSTRACT

The present contribution is chiefly a review, augmented by some new results on amphioxus and lamprey anatomy, that draws on paleontological and developmental data to suggest a scenario for cranial cartilage evolution in the phylum chordata. Consideration is given to the cartilage-related tissues of invertebrate chordates (amphioxus and some fossil groups like vetulicolians) as well as in the two major divisions of the subphylum Vertebrata (namely, agnathans, and gnathostomes). In the invertebrate chordates, which can be considered plausible proxy ancestors of the vertebrates, only a viscerocranium is present, whereas a neurocranium is absent. For this situation, we examine how cartilage-related tissues of this head region prefigure the cellular cartilage types in the vertebrates. We then focus on the vertebrate neurocranium, where cyclostomes evidently lack neural-crest derived trabecular cartilage (although this point needs to be established more firmly). In the more complex gnathostome, several neural-crest derived cartilage types are present: namely, the trabecular cartilages of the prechordal region and the parachordal cartilage the chordal region. In sum, we present an evolutionary framework for cranial cartilage evolution in chordates and suggest aspects of the subject that should profit from additional study.


Subject(s)
Lancelets , Vertebrates , Animals , Vertebrates/genetics , Skull , Cartilage , Neural Crest , Biological Evolution
3.
Cell Struct Funct ; 47(2): 55-73, 2022 Jul 27.
Article in English | MEDLINE | ID: mdl-35732428

ABSTRACT

The adherens junction (AJ) is an actin filament-anchoring junction. It plays a central role in epithelial morphogenesis through cadherin-based recognition and adhesion among cells. The stability and plasticity of AJs are required for the morphogenesis. An actin-binding α-catenin is an essential component of the cadherin-catenin complex and functions as a tension transducer that changes its conformation and induces AJ development in response to tension. Despite much progress in understanding molecular mechanisms of tension sensitivity of α-catenin, its significance on epithelial morphogenesis is still unknown. Here we show that the tension sensitivity of α-catenin is essential for epithelial cells to form round spheroids through proper multicellular rearrangement. Using a novel in vitro suspension culture model, we found that epithelial cells form round spheroids even from rectangular-shaped cell masses with high aspect ratios without using high tension and that increased tension sensitivity of α-catenin affected this morphogenesis. Analyses of AJ formation and cellular tracking during rounding morphogenesis showed cellular rearrangement, probably through AJ remodeling. The rearrangement occurs at the cell mass level, but not single-cell level. Hypersensitive α-catenin mutant-expressing cells did not show cellular rearrangement at the cell mass level, suggesting that the appropriate tension sensitivity of α-catenin is crucial for the coordinated round morphogenesis.Key words: α-catenin, vinculin, adherens junction, morphogenesis, mechanotransduction.


Subject(s)
Adherens Junctions , Mechanotransduction, Cellular , Adherens Junctions/metabolism , Cadherins , Morphogenesis , alpha Catenin/chemistry , alpha Catenin/metabolism
4.
Development ; 146(2)2019 01 16.
Article in English | MEDLINE | ID: mdl-30651295

ABSTRACT

Cell morphology changes dynamically during embryogenesis, and these changes create new interactions with surrounding cells, some of which are presumably mediated by intercellular signaling. However, the effects of morphological changes on intercellular signaling remain to be fully elucidated. In this study, we examined the effect of morphological changes in Wnt-producing cells on intercellular signaling in the spinal cord. After mid-gestation, roof-plate cells stretched along the dorsoventral axis in the mouse spinal cord, resulting in new contact at their tips with the ependymal cells that surround the central canal. Wnt1 and Wnt3a were produced by the stretched roof-plate cells and delivered to the cell process tip. Whereas Wnt signaling was activated in developing ependymal cells, Wnt activation in dorsal ependymal cells, which were close to the stretched roof plate, was significantly suppressed in embryos with roof plate-specific conditional knockout of Wls, which encodes a factor that is essential for Wnt secretion. Furthermore, proliferation of these cells was impaired in Wls conditional knockout mice during development and after induced spinal cord injury in adults. Therefore, morphological changes in Wnt-producing cells appear to generate new Wnt signal targets.


Subject(s)
Spinal Cord/pathology , Wnt Proteins/metabolism , Animals , Cell Proliferation , Embryo, Mammalian/pathology , Ependyma/embryology , Ependyma/pathology , Female , Ligands , Mice, Knockout , Morphogenesis , Pregnancy , Signal Transduction , Spinal Cord/embryology , Spinal Cord Injuries/embryology , Spinal Cord Injuries/pathology
5.
Proc Natl Acad Sci U S A ; 115(33): 8388-8393, 2018 08 14.
Article in English | MEDLINE | ID: mdl-30061390

ABSTRACT

The mechanosensory hair cells of the inner ear are required for hearing and balance and have a distinctive apical structure, the hair bundle, that converts mechanical stimuli into electrical signals. This structure comprises a single cilium, the kinocilium, lying adjacent to an ensemble of actin-based projections known as stereocilia. Hair bundle polarity depends on kinociliary protocadherin-15 (Pcdh15) localization. Protocadherin-15 is found only in hair-cell kinocilia, and is not localized to the primary cilia of adjacent supporting cells. Thus, Pcdh15 must be specifically targeted and trafficked into the hair-cell kinocilium. Here we show that kinocilial Pcdh15 trafficking relies on cell type-specific coupling to the generic intraflagellar transport (IFT) transport mechanism. We uncover a role for fibroblast growth factor receptor 1 (FGFR1) in loading Pcdh15 onto kinociliary transport particles in hair cells. We find that on activation, FGFR1 binds and phosphorylates Pcdh15. Moreover, we find a previously uncharacterized role for clathrin in coupling this kinocilia-specific cargo with the anterograde IFT-B complex through the adaptor, DAB2. Our results identify a modified ciliary transport pathway used for Pcdh15 transport into the cilium of the inner ear hair cell and coordinated by FGFR1 activity.


Subject(s)
Cadherins/physiology , Flagella/metabolism , Hair Cells, Auditory, Inner/metabolism , Protein Precursors/physiology , Receptor, Fibroblast Growth Factor, Type 1/physiology , Adaptor Proteins, Signal Transducing , Adaptor Proteins, Vesicular Transport/physiology , Animals , Apoptosis Regulatory Proteins , Cadherin Related Proteins , Chick Embryo , Clathrin/physiology , Mice , Phosphorylation , Protein Transport , Receptor, Fibroblast Growth Factor, Type 1/analysis
6.
Nature ; 505(7485): 676-80, 2014 Jan 30.
Article in English | MEDLINE | ID: mdl-24476891

ABSTRACT

We recently discovered an unexpected phenomenon of somatic cell reprogramming into pluripotent cells by exposure to sublethal stimuli, which we call stimulus-triggered acquisition of pluripotency (STAP). This reprogramming does not require nuclear transfer or genetic manipulation. Here we report that reprogrammed STAP cells, unlike embryonic stem (ES) cells, can contribute to both embryonic and placental tissues, as seen in a blastocyst injection assay. Mouse STAP cells lose the ability to contribute to the placenta as well as trophoblast marker expression on converting into ES-like stem cells by treatment with adrenocorticotropic hormone (ACTH) and leukaemia inhibitory factor (LIF). In contrast, when cultured with Fgf4, STAP cells give rise to proliferative stem cells with enhanced trophoblastic characteristics. Notably, unlike conventional trophoblast stem cells, the Fgf4-induced stem cells from STAP cells contribute to both embryonic and placental tissues in vivo and transform into ES-like cells when cultured with LIF-containing medium. Taken together, the developmental potential of STAP cells, shown by chimaera formation and in vitro cell conversion, indicates that they represent a unique state of pluripotency.


Subject(s)
Cell Differentiation , Cellular Reprogramming , Embryonic Stem Cells/cytology , Induced Pluripotent Stem Cells/cytology , Placenta/cytology , Trophoblasts/cytology , Adrenocorticotropic Hormone/pharmacology , Animals , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Lineage/drug effects , Cellular Reprogramming/drug effects , Embryonic Stem Cells/drug effects , Embryonic Stem Cells/metabolism , Epigenesis, Genetic/drug effects , Epigenesis, Genetic/genetics , Female , Fibroblast Growth Factor 4/pharmacology , Induced Pluripotent Stem Cells/drug effects , Leukemia Inhibitory Factor/pharmacology , Mice , Mice, Inbred ICR , Placenta/drug effects , Pregnancy , Trophoblasts/drug effects
7.
Development ; 143(20): 3806-3816, 2016 10 15.
Article in English | MEDLINE | ID: mdl-27578797

ABSTRACT

Signaling molecules have pleiotropic functions and are activated by various extracellular stimuli. Protein kinase C (PKC) is activated by diverse receptors, and its dysregulation is associated with diseases including cancer. However, how the undesired activation of PKC is prevented during development remains poorly understood. We have previously shown that a protein kinase, IKKε, is active at the growing bristle tip and regulates actin bundle organization during Drosophila bristle morphogenesis. Here, we demonstrate that IKKε regulates the actin bundle localization of a dynamic actin cross-linker, Fascin. IKKε inhibits PKC, thereby protecting Fascin from inhibitory phosphorylation. Excess PKC activation is responsible for the actin bundle defects in IKKε-deficient bristles, whereas PKC is dispensable for bristle morphogenesis in wild-type bristles, indicating that PKC is repressed by IKKε in wild-type bristle cells. These results suggest that IKKε prevents excess activation of PKC during bristle morphogenesis.


Subject(s)
Actins/metabolism , Carrier Proteins/metabolism , Drosophila Proteins/metabolism , Microfilament Proteins/metabolism , Protein Kinase C/metabolism , Actins/genetics , Animals , Carrier Proteins/genetics , Drosophila , Drosophila Proteins/genetics , I-kappa B Kinase/genetics , I-kappa B Kinase/metabolism , Microfilament Proteins/genetics , Phosphorylation , Protein Kinase C/genetics , Signal Transduction
8.
Genes Cells ; 23(5): 370-385, 2018 May.
Article in English | MEDLINE | ID: mdl-29542234

ABSTRACT

Mechanotransduction by α-catenin facilitates the force-dependent development of adherens junctions (AJs) by recruiting vinculin to reinforce actin anchoring of AJs. The α-catenin mechanotransducing action is facilitated by its force-sensing device region that autoinhibits the vinculin-binding site 1 (VBS1). Here, we report the high-resolution structure of the force-sensing device region of α-catenin, which shows the autoinhibited form comprised of helix bundles E, F and G. The cryptic VBS1 is embedded into helix bundle E stabilized by direct interactions with the autoinhibitory region forming helix bundles F and G. Our molecular dissection study showed that helix bundles F and G are stable in solution in each isolated form, whereas helix bundle E that contains VBS1 is unstable and intrinsically disordered in solution in the isolated form. We successfully identified key residues mediating the autoinhibition and produced mutated α-catenins that display variable force sensitivity and autoinhibition. Using these mutants, we demonstrate both in vitro and in vivo that, in the absence of this stabilization, the helix bundle containing VBS1 would adopt an unfolded form, thus exposing VBS for vinculin binding. We provide evidence for importance of mechanotransduction with the intrinsic force sensitivity for vinculin recruitment to adherens junctions of epithelial cell sheets with mutated α-catenins.


Subject(s)
Actins/metabolism , Adherens Junctions/physiology , Mechanotransduction, Cellular , Vinculin/metabolism , alpha Catenin/chemistry , alpha Catenin/metabolism , Animals , Binding Sites , Crystallography, X-Ray , Mice , Models, Molecular , Mutation , Protein Binding , Protein Conformation , Protein Folding , Vinculin/chemistry , Vinculin/genetics , alpha Catenin/genetics
9.
Proc Natl Acad Sci U S A ; 113(1): E81-90, 2016 Jan 05.
Article in English | MEDLINE | ID: mdl-26699487

ABSTRACT

Retinal transplantation therapy for retinitis pigmentosa is increasingly of interest due to accumulating evidence of transplantation efficacy from animal studies and development of techniques for the differentiation of human embryonic stem cells (hESCs) and induced pluripotent stem cells into retinal tissues or cells. In this study, we aimed to assess the potential clinical utility of hESC-derived retinal tissues (hESC-retina) using newly developed primate models of retinal degeneration to obtain preparatory information regarding the potential clinical utility of these hESC-retinas in transplantation therapy. hESC-retinas were first transplanted subretinally into nude rats with or without retinal degeneration to confirm their competency as a graft to mature to form highly specified outer segment structure and to integrate after transplantation. Two focal selective photoreceptor degeneration models were then developed in monkeys by subretinal injection of cobalt chloride or 577-nm optically pumped semiconductor laser photocoagulation. The utility of the developed models and a practicality of visual acuity test developed for monkeys were evaluated. Finally, feasibility of hESC-retina transplantation was assessed in the developed monkey models under practical surgical procedure and postoperational examinations. Grafted hESC-retina was observed differentiating into a range of retinal cell types, including rod and cone photoreceptors that developed structured outer nuclear layers after transplantation. Further, immunohistochemical analyses suggested the formation of host-graft synaptic connections. The findings of this study demonstrate the clinical feasibility of hESC-retina transplantation and provide the practical tools for the optimization of transplantation strategies for future clinical applications.


Subject(s)
Human Embryonic Stem Cells/cytology , Retina/cytology , Retina/transplantation , Retinal Degeneration/surgery , Animals , Cell Differentiation , Cobalt/toxicity , Disease Models, Animal , Haplorhini , Humans , Photoreceptor Cells/pathology , Primates , Rats , Retinal Degeneration/chemically induced , Retinal Degeneration/pathology
10.
Development ; 142(7): 1279-86, 2015 Apr 01.
Article in English | MEDLINE | ID: mdl-25742796

ABSTRACT

Birds and mammals, phylogenetically close amniotes with similar post-gastrula development, exhibit little conservation in their post-fertilization cleavage patterns. Data from the mouse suggest that cellular morphogenesis and molecular signaling at the cleavage stage play important roles in lineage specification at later (blastula and gastrula) stages. Very little is known, however, about cleavage-stage chick embryos, owing to their poor accessibility. This period of chick development takes place before egg-laying and encompasses several fundamental processes of avian embryology, including zygotic gene activation (ZGA) and blastoderm cell-layer increase. We have carried out morphological and cellular analyses of cleavage-stage chick embryos covering the first half of pre-ovipositional development, from Eyal-Giladi and Kochav stage (EGK-) I to EGK-V. Scanning electron microscopy revealed remarkable subcellular details of blastomere cellularization and subgerminal cavity formation. Phosphorylated RNA polymerase II immunostaining showed that ZGA in the chick starts at early EGK-III during the 7th to 8th nuclear division cycle, comparable with the time reported for other yolk-rich vertebrates (e.g. zebrafish and Xenopus). The increase in the number of cell layers after EGK-III is not a direct consequence of oriented cell division. Finally, we present evidence that, as in the zebrafish embryo, a yolk syncytial layer is formed in the avian embryo after EGK-V. Our data suggest that several fundamental features of cleavage-stage development in birds resemble those in yolk-rich anamniote species, revealing conservation in vertebrate early development. Whether this conservation lends morphogenetic support to the anamniote-to-amniote transition in evolution or reflects developmental plasticity in convergent evolution awaits further investigation.


Subject(s)
Cleavage Stage, Ovum/cytology , Embryonic Development , Vertebrates/embryology , Animals , Cell Nucleus/metabolism , Chick Embryo , Cleavage Stage, Ovum/ultrastructure , Egg Yolk/cytology , Embryo, Nonmammalian/cytology , Embryo, Nonmammalian/ultrastructure , Gene Expression Regulation, Developmental , Giant Cells/cytology , Mitosis , Phosphorylation , Phosphoserine/metabolism , RNA Polymerase II/metabolism , Zygote/metabolism
11.
Genes Cells ; 22(8): 723-741, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28639422

ABSTRACT

A spontaneous medaka ro mutant shows abnormal wobbling and rolling swimming behaviors. By positional cloning, we mapped the ro locus to a region containing the gene encoding Contactin1b (Cntn1b), which is an immunoglobulin (Ig)-superfamily domain-containing membrane-anchored protein. The ro mutant had a deletion in the cntn1b gene that introduced a premature stop codon. Furthermore, cntn1b mutants generated by the CRISPR/Cas9 system and trans-heterozygotes of the CRISPR mutant allele and ro had abnormal swimming behavior, indicating that the cntn1b gene was responsible for the ro-mutant phenotype. We also established zebrafish cntn1a and cntn1b mutants by transcription activator-like effector nucleases (TALENs). Zebrafish cntn1b but not cntn1a mutants showed abnormal swimming behaviors similar to those in the ro mutant, suggesting that Cntn1b plays a conserved role in the formation or function of the neural circuits that control swimming in teleosts. Although Cntn1-deficient mice have abnormal cerebellar neural circuitry, there was no apparent histological abnormality in the cerebellum of medaka or zebrafish cntn1b mutants. The medaka cntn1b mutants had defective optokinetic response (OKR) adaptation and abnormal rheotaxis (body positioning relative to water flow). Medaka and zebrafish cntn1b mutants are effective models for studying the neural circuits involved in motor learning and motor coordination.


Subject(s)
Codon, Terminator/genetics , Contactin 1/metabolism , Swimming , Zebrafish Proteins/metabolism , Animals , Cerebellum/metabolism , Cerebellum/physiology , Contactin 1/genetics , Learning , Motor Neurons/metabolism , Motor Neurons/physiology , Neural Pathways/metabolism , Neural Pathways/physiology , Oryzias , Zebrafish , Zebrafish Proteins/genetics
12.
PLoS Genet ; 11(10): e1005587, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26451951

ABSTRACT

Granule cells (GCs) are the major glutamatergic neurons in the cerebellum, and GC axon formation is an initial step in establishing functional cerebellar circuits. In the zebrafish cerebellum, GCs can be classified into rostromedial and caudolateral groups, according to the locations of their somata in the corresponding cerebellar lobes. The axons of the GCs in the caudolateral lobes terminate on crest cells in the dorsal hindbrain, as well as forming en passant synapses with Purkinje cells in the cerebellum. In the zebrafish mutant shiomaneki, the caudolateral GCs extend aberrant axons. Positional cloning revealed that the shiomaneki (sio) gene locus encodes Col4a6, a subunit of type IV collagen, which, in a complex with Col4a5, is a basement membrane (BM) component. Both col4a5 and col4a6 mutants displayed similar abnormalities in the axogenesis of GCs and retinal ganglion cells (RGCs). Although type IV collagen is reported to control axon targeting by regulating the concentration gradient of an axonal guidance molecule Slit, Slit overexpression did not affect the GC axons. The structure of the BM surrounding the tectum and dorsal hindbrain was disorganized in the col4a5 and col4a6 mutants. Moreover, the abnormal axogenesis of the caudolateral GCs and the RGCs was coupled with aberrant BM structures in the type IV collagen mutants. The regrowth of GC axons after experimental ablation revealed that the original and newly formed axons displayed similar branching and extension abnormalities in the col4a6 mutants. These results collectively suggest that type IV collagen controls GC axon formation by regulating the integrity of the BM, which provides axons with the correct path to their targets.


Subject(s)
Cerebellum/metabolism , Collagen Type IV/genetics , Retinal Ganglion Cells/metabolism , Zebrafish/genetics , Animals , Axons/metabolism , Basement Membrane/growth & development , Basement Membrane/metabolism , Cerebellum/growth & development , Collagen Type IV/metabolism , Extracellular Matrix/genetics , Extracellular Matrix/metabolism , Nerve Tissue Proteins/genetics , Purkinje Cells/metabolism , Zebrafish/growth & development , Zebrafish Proteins/genetics
13.
Dev Biol ; 415(1): 122-142, 2016 07 01.
Article in English | MEDLINE | ID: mdl-27174471

ABSTRACT

The processes of development leading up to gastrulation have been markedly altered during the evolution of amniotes, and it is uncertain how the mechanisms of axis formation are conserved and diverged between mouse and chick embryos. To assess the conservation and divergence of these mechanisms, this study examined gene expression patterns during the axis formation process in Chinese soft-shell turtle and Madagascar ground gecko preovipositional embryos. The data suggest that NODAL signaling, similarly to avian embryos but in contrast to eutherian embryos, does not have a role in epiblast and hypoblast development in reptilian embryos. The posterior marginal epiblast (PME) is the initial molecular landmark of axis formation in reptilian embryos prior to primitive plate development. Ontogenetically, PME may be the precursor of the primitive plate, and phylogenetically, Koller's sickle and posterior marginal zone in avian development may have been derived from the PME. Most of the genes expressed in the mouse anterior visceral endoderm (AVE genes), especially signaling antagonist genes, are not expressed in the hypoblast of turtle and gecko embryos, though they are expressed in the avian hypoblast. This study proposes that AVE gene expression in the hypoblast and the visceral endoderm could have been independently established in avian and eutherian lineages, similar to the primitive streak that has been independently acquired in these lineages.


Subject(s)
Body Patterning/physiology , Embryo, Nonmammalian/metabolism , Embryonic Development , Lizards/embryology , Turtles/embryology , Animals , Blastoderm/physiology , Body Patterning/genetics , Endoderm/metabolism , Gastrulation/physiology , Gene Expression Regulation, Developmental , Homeodomain Proteins/physiology , Lizards/genetics , Lizards/metabolism , Nodal Protein/physiology , Phylogeny , Primitive Streak/metabolism , Species Specificity , Transcription Factors/physiology , Turtles/genetics , Turtles/metabolism
14.
Nature ; 480(7375): 57-62, 2011 Nov 09.
Article in English | MEDLINE | ID: mdl-22080957

ABSTRACT

The adenohypophysis (anterior pituitary) is a major centre for systemic hormones. At present, no efficient stem-cell culture for its generation is available, partly because of insufficient knowledge about how the pituitary primordium (Rathke's pouch) is induced in the embryonic head ectoderm. Here we report efficient self-formation of three-dimensional adenohypophysis tissues in an aggregate culture of mouse embryonic stem (ES) cells. ES cells were stimulated to differentiate into non-neural head ectoderm and hypothalamic neuroectoderm in adjacent layers within the aggregate, and treated with hedgehog signalling. Self-organization of Rathke's-pouch-like three-dimensional structures occurred at the interface of these two epithelia, as seen in vivo, and various endocrine cells including corticotrophs and somatotrophs were subsequently produced. The corticotrophs efficiently secreted adrenocorticotropic hormone in response to corticotrophin releasing hormone and, when grafted in vivo, these cells rescued the systemic glucocorticoid level in hypopituitary mice. Thus, functional anterior pituitary tissue self-forms in ES cell culture, recapitulating local tissue interactions.


Subject(s)
Embryonic Stem Cells/cytology , Pituitary Gland, Anterior/cytology , Pituitary Gland, Anterior/embryology , Animals , Cell Culture Techniques , Cell Line , Cell Lineage , Cells, Cultured , Ectoderm/cytology , Ectoderm/embryology , Endocrine Cells/cytology , Endocrine Cells/metabolism , Hypopituitarism/pathology , Hypothalamus/cytology , Hypothalamus/embryology , Mice
15.
Proc Natl Acad Sci U S A ; 111(45): 16011-6, 2014 Nov 11.
Article in English | MEDLINE | ID: mdl-25355906

ABSTRACT

The cadherins Fat and Dachsous regulate cell polarity and proliferation via their heterophilic interactions at intercellular junctions. Their ectodomains are unusually large because of repetitive extracellular cadherin (EC) domains, which raises the question of how they fit in regular intercellular spaces. Cadherins typically exhibit a linear topology through the binding of Ca(2+) to the linker between the EC domains. Our electron-microscopic observations of mammalian Fat4 and Dachsous1 ectodomains, however, revealed that, although their N-terminal regions exhibit a linear configuration, the C-terminal regions are kinked with multiple hairpin-like bends. Notably, certain EC-EC linkers in Fat4 and Dachsous1 lost Ca(2+)-binding amino acids. When such non-Ca(2+)-binding linkers were substituted for a normal linker in E-cadherin, the mutant E-cadherins deformed more extensively than the wild-type molecule. To simulate cadherin structures with non-Ca(2+)-binding linkers, we used an elastic network model and confirmed that bent configurations can be generated by deformation of non-Ca(2+)-binding linkers. These findings suggest that Fat and Dachsous self-bend due to the loss of Ca(2+)-binding amino acids from specific EC-EC linkers, and can therefore adapt to confined spaces.


Subject(s)
Cadherins/metabolism , Calcium/metabolism , Intercellular Junctions/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Cadherin Related Proteins , Cadherins/genetics , HEK293 Cells , Humans , Intercellular Junctions/genetics , Intercellular Junctions/ultrastructure , Mice , Mutation , Protein Binding , Protein Structure, Tertiary , Tumor Suppressor Proteins/genetics
16.
Dev Dyn ; 245(12): 1176-1188, 2016 12.
Article in English | MEDLINE | ID: mdl-27666927

ABSTRACT

BACKGROUND: Previous comparative studies suggest that the requirement for Nodal in epiblast and hypoblast development is unique to mammalians. Expression of anterior visceral endoderm (AVE) genes in the visceral endoderm and of their orthologs in the hypoblast may be unique to mammalians and avians, and is absent in the reptilian hypoblast. Axis formation in reptiles is signaled by the formation of the posterior marginal epiblast (PME), which expresses a series of primitive streak genes. To assess the phylogenetic origin of Nodal and AVE gene expression and axis formation in amniotes, we examined marker gene expression in gray short-tailed opossum, a metatherian. RESULTS: Nodal was expressed in neither epiblast nor hypoblast of opossum embryos. No AVE genes were expressed in the opossum hypoblast. Attainment of polarity in the embryonic disk was signaled by Nodal, Wnt3a, Fgf8, and Bra expression in the PME at 8.5 days post-coitus. CONCLUSIONS: Nodal expression in epiblast or hypoblast may be unique to eutherians. AVE gene expression in visceral endoderm and hypoblast may have been independently acquired in eutherian and avian lineages. PME formation appears to be the event that signals axis formation in reptilian and metatherian embryos, and thus may be an ancestral characteristic of basal amniotes. Developmental Dynamics 245:1176-1188, 2016. © 2016 Wiley Periodicals, Inc.


Subject(s)
Monodelphis/embryology , Monodelphis/metabolism , Animals , Body Patterning/genetics , Body Patterning/physiology , Gene Expression Regulation, Developmental , Monodelphis/classification , Nodal Protein/genetics , Nodal Protein/metabolism , Phylogeny
17.
Dev Dyn ; 245(1): 67-86, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26404161

ABSTRACT

BACKGROUND: Mouse embryos are cup shaped, but most nonrodent eutherian embryos are disk shaped. Extraembryonic ectoderm (ExEc), which may have essential roles in anterior-posterior (A-P) axis formation in mouse embryos, does not develop in many eutherian embryos. To assess A-P axis formation in eutherians, comparative analyses were made on rabbit, porcine, and Suncus embryos. RESULTS: All embryos examined expressed Nodal initially throughout epiblast and visceral endoderm; its expression became restricted to the posterior region before gastrulation. Anterior visceral endoderm (AVE) genes were expressed in Otx2-positive visceral endoderm, with Dkk1 expression being most anterior. The mouse pattern of AVE formation was conserved in rabbit embryos, but had diverged in porcine and Suncus embryos. No structure that was molecularly equivalent to Bmp-positive ExEc, existed in rabbit or pig embryos. In Suncus embryos, A-P axis was determined at prehatching stage, and these embryos attached to uterine wall at future posterior side. CONCLUSIONS: Nodal, but not Bmp, functions in epiblast and visceral endoderm development may be conserved in eutherians. AVE functions may also be conserved, but the pattern of its formation has diverged among eutherians. Roles of BMP and NODAL gradients in AVE formation seem to have been established in a subset of rodents.


Subject(s)
Ectoderm/physiology , Embryonic Development/physiology , Endoderm/physiology , Gene Expression Regulation, Developmental , Animals , Body Patterning/genetics , Intercellular Signaling Peptides and Proteins/genetics , Nodal Protein/genetics , Rabbits , Swine
18.
J Cell Sci ; 127(Pt 9): 2040-52, 2014 May 01.
Article in English | MEDLINE | ID: mdl-24610943

ABSTRACT

Cdc42 is a key regulator of dynamic actin organization. However, little is known about how Cdc42-dependent actin regulation influences steady-state actin structures in differentiated epithelia. We employed inner ear hair-cell-specific conditional knockout to analyze the role of Cdc42 in hair cells possessing highly elaborate stable actin protrusions (stereocilia). Hair cells of Atoh1-Cre;Cdc42(flox/flox) mice developed normally but progressively degenerated after maturation, resulting in progressive hearing loss particularly at high frequencies. Cochlear hair cell degeneration was more robust in inner hair cells than in outer hair cells, and began as stereocilia fusion and depletion, accompanied by a thinning and waving circumferential actin belt at apical junctional complexes (AJCs). Adenovirus-encoded GFP-Cdc42 expression in hair cells and fluorescence resonance energy transfer (FRET) imaging of hair cells from transgenic mice expressing a Cdc42-FRET biosensor indicated Cdc42 presence and activation at stereociliary membranes and AJCs in cochlear hair cells. Cdc42-knockdown in MDCK cells produced phenotypes similar to those of Cdc42-deleted hair cells, including abnormal microvilli and disrupted AJCs, and downregulated actin turnover represented by enhanced levels of phosphorylated cofilin. Thus, Cdc42 influenced the maintenance of stable actin structures through elaborate tuning of actin turnover, and maintained function and viability of cochlear hair cells.


Subject(s)
Hair Cells, Auditory/metabolism , cdc42 GTP-Binding Protein/metabolism , Actins/metabolism , Animals , Biosensing Techniques , Cochlea/cytology , Cochlea/metabolism , Dogs , Fluorescence Resonance Energy Transfer , Humans , Immunohistochemistry , In Situ Hybridization , Madin Darby Canine Kidney Cells , Mice , Microscopy, Electrochemical, Scanning , Microscopy, Electron, Transmission , Organ Culture Techniques/methods , cdc42 GTP-Binding Protein/genetics
19.
Cell Tissue Res ; 366(1): 75-87, 2016 10.
Article in English | MEDLINE | ID: mdl-27177867

ABSTRACT

Shootin1 is a brain-specific cytoplasmic protein involved in neuronal polarity formation and axon outgrowth. It accumulates at the leading edge of axonal growth cones, where it mediates the mechanical coupling between F-actin retrograde flow and cell adhesions as a clutch molecule, thereby producing force for axon outgrowth. In this study, we report a novel splicing isoform of shootin1 which is expressed not only in the brain but also in peripheral tissues. We have renamed the brain-specific shootin1 as shootin1a and termed the novel isoform as shootin1b. Immunoblot and immunohistochemical analyses with a shootin1b-specific antibody revealed that shootin1b is distributed in various mouse tissues including the lung, liver, stomach, intestines, spleen, pancreas, kidney and skin. Interestingly, shootin1b immunoreactivity was widely detected in epithelial cells that constitute simple and stratified epithelia; in some cells, it colocalized with E-cadherin and cortactin at cell-cell contact sites. Shootin1b also localized in dendritic cells in the spleen. These results suggest that shootin1b may function in various peripheral tissues including epithelial cells.


Subject(s)
Nerve Tissue Proteins/metabolism , Organ Specificity , Aging/metabolism , Amino Acid Sequence , Animals , Antibody Specificity , Cadherins/metabolism , Cell Communication , Cortactin/metabolism , Immunohistochemistry , Mice, Inbred C57BL , Nerve Tissue Proteins/chemistry , Protein Isoforms , Protein Transport , Rats , Tissue Distribution
20.
FASEB J ; 29(5): 2137-49, 2015 May.
Article in English | MEDLINE | ID: mdl-25678623

ABSTRACT

Endoplasmic reticulum (ER) stress is a cellular condition in which unfolded proteins accumulate in the ER because of various but specific causes. Physiologic ER stress occurs transiently during myoblast differentiation, and although its cause remains unknown, it plays a critical role in myofiber formation. To examine the mechanism underlying ER stress, we monitored ER morphology during differentiation of murine myoblasts. Novel ER-derived structures transiently appeared prior to myoblast fusion both in vitro and in vivo. Electron microscopy studies revealed that these structures consisted of pseudoconcentric ER cisternae with narrow lumens. Similar structures specifically formed by pharmacologically induced ER Ca(2+) depletion, and inhibition of ER Ca(2+) efflux channels in differentiating myoblasts considerably suppressed ER-specific deformation and ER stress signaling. Thus, we named the novel structures stress-activated response to Ca(2+) depletion (SARC) bodies. Prior to SARC body formation, stromal interaction molecule 1 (STIM1), an ER Ca(2+) sensor protein, formed ER Ca(2+) depletion-specific clusters. Furthermore, myoblast differentiation manifested by myoblast fusion did not proceed under the same conditions as inhibition of ER Ca(2+) depletion. Altogether, these observations suggest that ER Ca(2+) depletion is a prerequisite for myoblast fusion, causing both physiologic ER stress signaling and SARC body formation.


Subject(s)
Calcium/metabolism , Cell Differentiation , Cell Membrane/metabolism , Endoplasmic Reticulum Stress , Endoplasmic Reticulum/metabolism , Myoblasts, Skeletal/cytology , Animals , Blotting, Western , Calcium Channels/metabolism , Cells, Cultured , Immunoenzyme Techniques , Mice , Myoblasts, Skeletal/metabolism , Signal Transduction , Stromal Interaction Molecule 1
SELECTION OF CITATIONS
SEARCH DETAIL