Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
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
2.
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, IKKƎĀµ, is active at the growing bristle tip and regulates actin bundle organization during Drosophila bristle morphogenesis. Here, we demonstrate that IKKƎĀµ regulates the actin bundle localization of a dynamic actin cross-linker, Fascin. IKKƎĀµ inhibits PKC, thereby protecting Fascin from inhibitory phosphorylation. Excess PKC activation is responsible for the actin bundle defects in IKKƎĀµ-deficient bristles, whereas PKC is dispensable for bristle morphogenesis in wild-type bristles, indicating that PKC is repressed by IKKƎĀµ in wild-type bristle cells. These results suggest that IKKƎĀµ 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
3.
Proc Natl Acad Sci U S A ; 113(2): 332-7, 2016 Jan 12.
Article in English | MEDLINE | ID: mdl-26715742

ABSTRACT

Polarized epithelial cells exhibit a characteristic array of microtubules that are oriented along the apicobasal axis of the cells. The minus-ends of these microtubules face apically, and the plus-ends face toward the basal side. The mechanisms underlying this epithelial-specific microtubule assembly remain unresolved, however. Here, using mouse intestinal cells and human Caco-2 cells, we show that the microtubule minus-end binding protein CAMSAP3 (calmodulin-regulated-spectrin-associated protein 3) plays a pivotal role in orienting the apical-to-basal polarity of microtubules in epithelial cells. In these cells, CAMSAP3 accumulated at the apical cortices, and tethered the longitudinal microtubules to these sites. Camsap3 mutation or depletion resulted in a random orientation of these microtubules; concomitantly, the stereotypic positioning of the nucleus and Golgi apparatus was perturbed. In contrast, the integrity of the plasma membrane was hardly affected, although its structural stability was decreased. Further analysis revealed that the CC1 domain of CAMSAP3 is crucial for its apical localization, and that forced mislocalization of CAMSAP3 disturbs the epithelial architecture. These findings demonstrate that apically localized CAMSAP3 determines the proper orientation of microtubules, and in turn that of organelles, in mature mammalian epithelial cells.


Subject(s)
Cell Polarity , Epithelial Cells/cytology , Epithelial Cells/metabolism , Microtubule-Associated Proteins/metabolism , Microtubules/metabolism , Organelles/metabolism , Amino Acid Sequence , Animals , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Caco-2 Cells , Enterocytes/cytology , Enterocytes/metabolism , Enterocytes/ultrastructure , Epithelial Cells/ultrastructure , Green Fluorescent Proteins/metabolism , Homozygote , Humans , Mice, Inbred C57BL , Mice, Mutant Strains , Microtubule-Associated Proteins/chemistry , Models, Biological , Molecular Sequence Data , Mutation/genetics , Nocodazole/pharmacology , Protein Structure, Tertiary , Subcellular Fractions/metabolism , Thiazolidines/pharmacology
4.
Development ; 138(21): 4621-6, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21989911

ABSTRACT

Joints permit efficient locomotion, especially among animals with a rigid skeleton. Joint morphologies vary in the body of individual animals, and the shapes of homologous joints often differ across species. The diverse locomotive behaviors of animals are based, in part, on the developmental and evolutionary history of joint morphogenesis. We showed previously that strictly coordinated cell-differentiation and cell-movement events within the epidermis sculpt the interlocking ball-and-socket joints in the adult Drosophila tarsus (distal leg). Here, we show that the tarsal joints of various insect species can be classified into three types: ball-and-socket, side-by-side and uniform. The last two probably result from joint formation without the cell-differentiation step, the cell-movement step, or both. Similar morphological variations were observed in Drosophila legs when Notch function was temporarily blocked during joint formation, implying that the independent acquisition of cell differentiation and cell movement underlay the elaboration of tarsal joint morphologies during insect evolution. These results provide a framework for understanding how the seemingly complex morphology of the interlocking joint could have developed during evolution by the addition of simple developmental modules: cell differentiation and cell movement.


Subject(s)
Arthropods/anatomy & histology , Biological Evolution , Drosophila Proteins/metabolism , Drosophila/anatomy & histology , Insecta/anatomy & histology , Phenotype , Receptors, Notch/metabolism , Animals , Arthropods/classification , Arthropods/physiology , Cell Differentiation/physiology , Drosophila/classification , Drosophila/physiology , Drosophila Proteins/genetics , Extremities , Insecta/classification , Insecta/physiology , Joints/anatomy & histology , Joints/physiology , Receptors, Notch/genetics
5.
Development ; 137(12): 2055-63, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20501594

ABSTRACT

Animal body shape is framed by the skeleton, which is composed of extracellular matrix (ECM). Although how the body plan manifests in skeletal morphology has been studied intensively, cellular mechanisms that directly control skeletal ECM morphology remain elusive. In particular, how dynamic behaviors of ECM-secreting cells, such as shape changes and movements, contribute to ECM morphogenesis is unclear. Strict control of ECM morphology is crucial in the joints, where opposing sides of the skeleton must have precisely reciprocal shapes to fit each other. Here we found that, in the development of ball-and-socket joints in the Drosophila leg, the two sides of ECM form sequentially. We show that distinct cell populations produce the 'ball' and the 'socket', and that these cells undergo extensive shape changes while depositing ECM. We propose that shape changes of ECM-producing cells enable the sequential ECM formation to allow the morphological coupling of adjacent components. Our results highlight the importance of dynamic cell behaviors in precise shaping of skeletal ECM architecture.


Subject(s)
Drosophila/physiology , Extracellular Matrix/physiology , Extremities/growth & development , Joints/growth & development , Morphogenesis , Animals
6.
Sci Rep ; 11(1): 5857, 2021 03 12.
Article in English | MEDLINE | ID: mdl-33712686

ABSTRACT

Epithelial cells organize an ordered array of non-centrosomal microtubules, the minus ends of which are regulated by CAMSAP3. The role of these microtubules in epithelial functions, however, is poorly understood. Here, we show that the kidneys of mice in which Camsap3 is mutated develop cysts at the proximal convoluted tubules (PCTs). PCTs were severely dilated in the mutant kidneys, and they also exhibited enhanced cell proliferation. In these PCTs, epithelial cells became flattened along with perturbation of microtubule arrays as well as of certain subcellular structures such as interdigitating basal processes. Furthermore, YAP and PIEZO1, which are known as mechanosensitive regulators for cell shaping and proliferation, were activated in these mutant PCT cells. These observations suggest that CAMSAP3-mediated microtubule networks are important for maintaining the proper mechanical properties of PCT cells, and its loss triggers cell deformation and proliferation via activation of mechanosensors, resulting in the dilation of PCTs.


Subject(s)
Cysts/pathology , Kidney Tubules, Proximal/metabolism , Kidney Tubules, Proximal/pathology , Microtubule-Associated Proteins/metabolism , Microtubules/metabolism , Animals , Cell Proliferation , Cysts/physiopathology , Epithelial Cells/metabolism , Epithelial Cells/pathology , Epithelial Cells/ultrastructure , Ion Channels/metabolism , Kidney Tubules, Proximal/physiopathology , Kidney Tubules, Proximal/ultrastructure , Mice, Knockout , Mice, Mutant Strains , Myosins/metabolism , YAP-Signaling Proteins/metabolism
7.
Mol Biol Cell ; 32(20): ar12, 2021 10 01.
Article in English | MEDLINE | ID: mdl-34319756

ABSTRACT

Motile cilia of multiciliated epithelial cells undergo synchronized beating to produce fluid flow along the luminal surface of various organs. Each motile cilium consists of an axoneme and a basal body (BB), which are linked by a "transition zone" (TZ). The axoneme exhibits a characteristic 9+2 microtubule arrangement important for ciliary motion, but how this microtubule system is generated is not yet fully understood. Here we show that calmodulin-regulated spectrin-associated protein 3 (CAMSAP3), a protein that can stabilize the minus-end of a microtubule, concentrates at multiple sites of the cilium-BB complex, including the upper region of the TZ or the axonemal basal plate (BP) where the central pair of microtubules (CP) initiates. CAMSAP3 dysfunction resulted in loss of the CP and partial distortion of the BP, as well as the failure of multicilia to undergo synchronized beating. These findings suggest that CAMSAP3 plays pivotal roles in the formation or stabilization of the CP by localizing at the basal region of the axoneme and thereby supports the coordinated motion of multicilia in airway epithelial cells.


Subject(s)
Cilia/metabolism , Microtubule-Associated Proteins/metabolism , Microtubules/metabolism , Actin Cytoskeleton/metabolism , Animals , Axoneme/physiology , Basal Bodies/physiology , Epithelial Cells/metabolism , Female , Male , Mice , Mice, Inbred ICR , Mice, Transgenic , Movement/physiology , Trachea/physiology
8.
Plant J ; 57(5): 819-31, 2009 Mar.
Article in English | MEDLINE | ID: mdl-18980648

ABSTRACT

The preprophase band (PPB) marks the site on the plant cell cortex where the cell plate will fuse during the final stage of cytokinesis. Recent studies have shown that several cytoskeletal proteins are depleted at the PPB site, but the processes that bring about these changes are still unknown. We have investigated the membrane systems associated with the PPB regions of epidermal cells of onion cotyledons by means of serial thin sections and electron tomograms. In contrast with specimens preserved by chemical fixatives, our high-pressure frozen cells demonstrated the presence of large numbers of clathrin-coated pits and vesicles in the PPB regions. The vesicles were of two types: clathrin-coated and structurally related, non-coated vesicles. Quantitative analysis of the data revealed that the number of clathrin-coated pits and vesicles is higher in the PPB regions than outside of these regions. Immunofluorescent microscopy using anti-plant clathrin-antibody confirmed this result. In contrast, no differences in secretory activities were observed. We postulate that the removal of membrane proteins by endocytosis plays a role in the formation of PPB 'memory' structures.


Subject(s)
Clathrin/metabolism , Endocytosis , Onions/growth & development , Plant Epidermis/growth & development , Prophase , Clathrin-Coated Vesicles/metabolism , Coated Pits, Cell-Membrane/metabolism , Coated Pits, Cell-Membrane/ultrastructure , Cytokinesis , Electron Microscope Tomography , Onions/cytology , Plant Epidermis/cytology , Plant Epidermis/ultrastructure
9.
Genes Cells ; 14(3): 381-93, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19210548

ABSTRACT

C-terminal Src kinase (Csk) is a non-receptor type of tyrosine kinase, and serves as an essential negative regulator of Src family tyrosine kinases (SFKs) in vertebrates. However, analyses of Csk and SFKs from primitive animals suggest that the Csk-mediated mechanisms regulating SFK activity might diverge between evolutional branches, different tissues or SFK family members. We examined in vivo roles of CSK-1, a Caenorhabditis elegans orthologue of Csk, by generating animals lacking csk-1 function. Although some csk-1 mutants died during embryogenesis, the majority of mutants died during the first stage of larval development. In csk-1 mutants, the function of pharyngeal muscles, the major site of CSK-1 expression, was severely damaged. The pumping of pharyngeal grinder cells became arrhythmic, causing disabled feeding. Electron microscopy showed that pharyngeal muscle filaments were disorientated in the csk-1 mutants. These indicate that CSK-1 is crucial for proper organization of pharyngeal muscles. However, the growth arrest phenotype in csk-1 mutants could not be suppressed by src-1 and/or src-2 mutation, and SRC-1 was not significantly activated in the csk-1 mutants. These results suggest that CSK-1 has an essential function in organization of pharyngeal muscle filaments that does not require C. elegans SFKs.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/metabolism , Protein Kinases/metabolism , Animals , CSK Tyrosine-Protein Kinase , Caenorhabditis elegans/embryology , Caenorhabditis elegans Proteins/genetics , Embryo, Nonmammalian/metabolism , Gene Deletion , Pharyngeal Muscles/embryology , Pharyngeal Muscles/metabolism , Protein Kinases/genetics
10.
Curr Biol ; 29(9): 1512-1520.e6, 2019 05 06.
Article in English | MEDLINE | ID: mdl-31006566

ABSTRACT

Nanometer-level patterned surface structures form the basis of biological functions, including superhydrophobicity, structural coloration, and light absorption [1-3]. In insects, the cuticle overlying the olfactory sensilla has multiple small (50- to 200-nm diameter) pores [4-8], which are supposed to function as a filter that admits odorant molecules, while preventing the entry of larger airborne particles andĀ limiting water loss. However, the cellular processes underlying the patterning of extracellular matrices into functional nano-structures remain unknown. Here, we show that cuticular nanopores in Drosophila olfactory sensilla originate from a curved ultrathin film that is formed in the outermost envelope layer of the cuticle and secreted from specialized protrusions in the plasma membrane of the hair forming (trichogen) cell. The envelope curvature coincides with plasma membrane undulations associated with endocytic structures. The gore-tex/Osiris23 gene encodes an endosomal protein that is essential for envelope curvature, nanopore formation, and odor receptivity and is expressed specifically in developing olfactory trichogen cells. The 24-member Osiris gene family is expressed in cuticle-secreting cells and is found only in insect genomes. These results reveal an essential requirement for nanopores for odor reception and identify Osiris genes as a platform for investigating the evolution of surface nano-fabrication in insects.


Subject(s)
Drosophila melanogaster/ultrastructure , Sensilla/ultrastructure , Animals , Female , Microscopy, Electron, Transmission , Nanopores/ultrastructure
11.
Elife ; 72018 04 03.
Article in English | MEDLINE | ID: mdl-29609731

ABSTRACT

VEGF secreted from retinal pigment epithelial (RPE) cells is responsible for the choroidal vascular development; however, the molecular regulatory mechanism is unclear. We found that Aldh1a1-/- mice showed choroidal hypoplasia with insufficient vascularization in the dorsal region, although Aldh1a1, an enzyme that synthesizes retinoic acids (RAs), is expressed in the dorsal neural retina, not in the RPE/choroid complex. The level of VEGF in the RPE/choroid was significantly decreased in Aldh1a1-/- mice, and RA-dependent enhancement of VEGF was observed in primary RPE cells. An RA-deficient diet resulted in dorsal choroidal hypoplasia, and simple RA treatment of Aldh1a1-/- pregnant females suppressed choroid hypoplasia in their offspring. We also found downregulation of Sox9 in the dorsal neural retina and RPE of Aldh1a1-/- mice and RPE-specific disruption of Sox9 phenocopied Aldh1a1-/- choroidal development. These results suggest that RAs produced by Aldh1a1 in the neural retina directs dorsal choroidal vascular development via Sox9 upregulation in the dorsal RPE cells to enhance RPE-derived VEGF secretion.


Subject(s)
Aldehyde Dehydrogenase/metabolism , Choroid/physiology , Gene Expression Regulation , Neovascularization, Physiologic , Retinal Pigment Epithelium/physiology , SOX9 Transcription Factor/metabolism , Vascular Endothelial Growth Factor A/metabolism , Aldehyde Dehydrogenase/genetics , Aldehyde Dehydrogenase 1 Family , Animals , Epithelial Cells/physiology , Mice, Inbred C57BL , Mice, Knockout , Retinal Dehydrogenase , Tretinoin/metabolism
12.
Dev Cell ; 37(3): 226-37, 2016 05 09.
Article in English | MEDLINE | ID: mdl-27165555

ABSTRACT

Differentiation of pluripotent stem cells (PSCs) requires transposon silencing throughout the process. PIWIs, best known as key factors in germline transposon silencing, are also known to act in somatic differentiation of planarian PSCs (neoblasts). However, how PIWIs control the latter process remains elusive. Here, using Dugesia japonica, we show that a nuclear PIWI, DjPiwiB, was bound to PIWI-interacting RNAs (generally key mediators of PIWI-dependent transposon silencing), and was detected in not only neoblasts but also their descendant somatic cells, which do not express piwi. In contrast, cytoplasmic DjPiwiA and DjPiwiC were detected only in neoblasts, in accord with their transcription there. DjPiwiB was indispensable for regeneration, but dispensable for transposon silencing in neoblasts. However, transposons were derepressed at the onset of differentiation in DjPiwiB-knockdown planarians. Thus, DjPiwiB appears to be inherited by descendant somatic cells of neoblasts to ensure transposon silencing in those cells, which are unable to produce PIWI proteins.


Subject(s)
Argonaute Proteins/metabolism , Cell Differentiation , Cell Nucleus/metabolism , DNA Transposable Elements/genetics , Inheritance Patterns/genetics , Planarians/cytology , Planarians/genetics , Pluripotent Stem Cells/metabolism , Animals , Base Sequence , Gene Silencing , Immunohistochemistry , Models, Biological , RNA, Small Interfering/metabolism
13.
Mol Biol Cell ; 27(11): 1809-20, 2016 06 01.
Article in English | MEDLINE | ID: mdl-27053663

ABSTRACT

The preprophase band (PPB) is a cytokinetic apparatus that determines the site of cell division in plants. It originates as a broad band of microtubules (MTs) in G2 and narrows to demarcate the future division site during late prophase. Studies with fluorescent probes have shown that PPBs contain F-actin during early stages of their development but become actin depleted in late prophase. Although this suggests that actins contribute to the early stages of PPB formation, how actins contribute to PPB-MT organization remains unsolved. To address this question, we used electron tomography to investigate the spatial relationship between microfilaments (MFs) and MTs at different stages of PPB assembly in onion cotyledon epidermal cells. We demonstrate that the PPB actins observed by fluorescence microscopy correspond to short, single MFs. A majority of the MFs are bound to MTs, with a subset forming MT-MF-MT bridging structures. During the later stages of PPB assembly, the MF-mediated links between MTs are displaced by MT-MT linkers as the PPB MT arrays mature into tightly packed MT bundles. On the basis of these observations, we propose that the primary function of actins during PPB formation is to mediate the initial bundling of the PPB MTs.


Subject(s)
Actin Cytoskeleton/metabolism , Cotyledon/metabolism , Microtubules/metabolism , Onions/metabolism , Actins/metabolism , Cell Division/physiology , Cotyledon/cytology , Cytokinesis , Cytoskeleton/metabolism , Electron Microscope Tomography , Mitosis , Onions/cytology , Prophase , Tubulin/metabolism
14.
Brain Res Dev Brain Res ; 159(2): 127-34, 2005 Oct 06.
Article in English | MEDLINE | ID: mdl-16139369

ABSTRACT

The cytoarchitecture of dorsal cochlear nucleus (DCN), characterized by a distinct laminar structure similar to the cerebellar cortex of the normal mouse, is known to be disrupted in the Reelin-deficient mouse, reeler. Here, we have reexamined both the cytoarchitecture and myeloarchitecture of this nucleus and described expression pattern of Reelin protein during perinatal periods. Reelin-immunopositive granule cells were firstly recognized in the external granular layer of the DCN at embryological day 16 (E16). Next, we examined the cytoarchitecture of the DCN of the normal and reeler mice with Ca2+/calmodulin-dependent protein kinase IIalpha (CaMKIIalpha) immunostaining. CaMKIIalpha-immunoreactive cartwheel cells were laminarly distributed in the layer II of the normal DCN, but scattered throughout the reeler DCN. Injection of retrograde tracer, Fluoro-Gold (FG) into the inferior colliculus of the reeler mouse resulted in that retrogradely labeled neurons in the DCN were radially scattered instead of being confined to a single layer as seen in the normal mouse. To examine whether CaMKIIalpha-immunopositive cartwheel cells are neurons projecting to the inferior colliculus or not, double labeling with CaMKIIalpha immunohistochemistry and retrograde labeling with an injection of FG into the inferior colliculus were made, which revealed that CaMKIIalpha-immunoreactive cartwheel cells do not send axons to the inferior colliculus. The present findings imply that Reelin may have some roles in the formation of laminar structures of the DCN.


Subject(s)
Auditory Pathways/cytology , Cell Adhesion Molecules, Neuronal/biosynthesis , Cochlear Nucleus/cytology , Cochlear Nucleus/metabolism , Cochlear Nucleus/pathology , Extracellular Matrix Proteins/biosynthesis , Nerve Tissue Proteins/biosynthesis , Serine Endopeptidases/biosynthesis , Animals , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cell Adhesion Molecules, Neuronal/genetics , Extracellular Matrix Proteins/genetics , Immunohistochemistry , In Situ Hybridization , Mice , Mice, Mutant Strains , Mutation , Nerve Tissue Proteins/genetics , Neurons/metabolism , Neurons/pathology , RNA, Messenger/analysis , Reelin Protein , Serine Endopeptidases/genetics
15.
Dev Cell ; 35(2): 236-46, 2015 Oct 26.
Article in English | MEDLINE | ID: mdl-26506310

ABSTRACT

Determination of left-right asymmetry in mouse embryos is established by a leftward fluid flow that is generated by clockwise rotation of node cilia. How node cilia achieve stable unidirectional rotation has remained unknown, however. Here we show that brief exposure to the microtubule-stabilizing drug paclitaxel (Taxol) induces randomly directed rotation and changes the ultrastructure of node cilia. In vivo observations and a computer simulation revealed that a regular 9+0 arrangement of doublet microtubules is essential for stable unidirectional rotation of node cilia. The 9+2 motile cilia of the airway, which manifest planar beating, are resistant to Taxol treatment. However, the airway cilia of mice lacking the radial spoke head protein Rsph4a undergo rotational movement instead of planar beating, are prone to microtubule rearrangement, and are sensitive to Taxol. Our results suggest that the absence of radial spokes allows node cilia to rotate unidirectionally but, as a trade-off, renders them ultrastructurally fragile.


Subject(s)
Body Patterning/genetics , Cilia/genetics , Embryonic Development/genetics , Animals , Cilia/physiology , Cilia/ultrastructure , Embryo, Mammalian , Embryonic Development/drug effects , Mice , Microtubules/drug effects , Microtubules/ultrastructure , Paclitaxel/administration & dosage
16.
J Comp Neurol ; 463(3): 303-15, 2003 Aug 25.
Article in English | MEDLINE | ID: mdl-12820163

ABSTRACT

The shaking rat Kawasaki (SRK) is an autosomal recessive mutant that exhibits reeler-like abnormal locomotor behaviors. The murine reeler mutants arise from several mutations in the specific gene called reelin, which result in defects of Reelin expression or secretion in the cerebral cortex and other regions of CNS. To address the issue of whether the SRK mutation also arises from a mutation in reelin, we analyzed the reelin gene in SRK. Northern analysis of reelin mRNA from normal rats showed that rat reelin was expressed as a approximately 12-kb transcript in both the cerebrum and the cerebellum, whereas reelin expression was markedly reduced in the SRK brains. In situ hybridization analysis showed that reelin mRNA in the SRK brains was expressed in Cajal-Retzius cells in the marginal zone of the cerebral cortex and outer granular cells in the cerebellar cortex in similar manners to normal controls, but its expression was considerably reduced. On Western blotting and immunohistochemical analyses using antibodies specific for the Reelin protein, no immunoproduct was recognized in the cerebral and cerebellar cortices. From the cDNA sequences, we found a 64-base heterologous sequence in SRK reelin, which contains a termination codon in the reading frame. Furthermore, genomic DNA analysis revealed that a 10-base deletion, which contains a predicted splice donor site, occurred in the SRK genomic reelin gene, resulting in "read through" into the following intron in SRK. Thus, the SRK mutation is another type of mutation that lacks expression of the functional Reelin protein and, therefore, causes the reeler phenotype.


Subject(s)
Brain/physiology , Cell Adhesion Molecules, Neuronal/genetics , DNA, Complementary , Extracellular Matrix Proteins/genetics , Gait Disorders, Neurologic/genetics , Animals , Base Sequence , Blotting, Northern , Blotting, Western , DNA Mutational Analysis , DNA, Complementary/analysis , Gene Deletion , Immunohistochemistry , In Situ Hybridization , Molecular Sequence Data , Mutation , Nerve Tissue Proteins , Neurons/physiology , RNA, Messenger/analysis , Rats , Rats, Mutant Strains , Rats, Wistar , Reelin Protein , Serine Endopeptidases
17.
Brain Res Dev Brain Res ; 148(1): 89-96, 2004 Jan 31.
Article in English | MEDLINE | ID: mdl-14757522

ABSTRACT

Reelin is an extracellular matrix protein, which plays a crucial role for the formation of laminated and nonlaminated structures in the central nervous system. To elucidate its roles in the postnatal brain, in the present study, we raised a polyclonal antibody specific for rat Reelin, and investigated Reelin-expressing neurons in the rat brain during the postnatal periods in detail. We found that some Reelin-expressing cells existed in the anterior commissure and corpus callosum. These Reelin-expressing cells were also immunostained with the antibody specific for neurons, but not immunostained with the antibodies specific for astrocytes nor oligodendrocytes, suggesting that these Reelin-expressing cells in the white matter are neurons. They are also immunostained with anti-GAD67 antibody, indicating that Reelin-expressing cells in the commissure systems are GABAergic neurons. Reelin-expressing neurons in the anterior commissure had many conspicuous varicosities on their dendritic arbors and mimic to the interfascicular neurons. These results suggest that Reelin may participate in the regulatory mechanism of neuronal activities through the commissure structure during the postnatal periods.


Subject(s)
Cell Adhesion Molecules, Neuronal/metabolism , Corpus Callosum/cytology , Extracellular Matrix Proteins/metabolism , Neurons/metabolism , Septum of Brain/cytology , Animals , Animals, Genetically Modified , Animals, Newborn , Blotting, Western/methods , Cell Adhesion Molecules, Neuronal/genetics , Corpus Callosum/growth & development , Corpus Callosum/metabolism , Extracellular Matrix Proteins/genetics , Glutamate Decarboxylase/metabolism , Immunohistochemistry/methods , Isoenzymes/metabolism , Mice , Mice, Inbred C57BL , Mice, Neurologic Mutants , Nerve Tissue Proteins , Oligodendroglia/metabolism , Phosphopyruvate Hydratase/metabolism , Rats , Rats, Wistar , Reelin Protein , Septum of Brain/growth & development , Septum of Brain/metabolism , Serine Endopeptidases
18.
Brain Res Dev Brain Res ; 141(1-2): 1-13, 2003 Mar 14.
Article in English | MEDLINE | ID: mdl-12644243

ABSTRACT

Shaking Rat Kawasaki (SRK) is a Reelin-deficient rat, that shows significant cytoarchitectural abnormalities in the cerebral and cerebellar cortices in a similar manner to the reeler malformation. In the present study, we investigated the cytoarchitecture and myeloarchitecture of the superior colliculus (SC) of this mutant rat. The Nissl staining clearly showed that neuronal components in the superficial layers of the SC in SRK rat were intermingled with each other and that the boundaries between these superficial layers were blurred. The MBP immunohistochemistry showed an abnormal fiber pattern in the superficial layers of the SC of this mutant rat. In the normal rat, myelinated fibers passed rostrocaudally through the optic layer, and only a few myelinated fibers were recognized in the uppermost two layers, i.e., the zonal and superficial gray layers. By contrast, in SRK rat, the myelinated fibers were distributed throughout the entire thickness of the superficial layers of the SC. Anterograde labeling of retinotectal fibers with an injection of Cholera Toxin subunit B into the retina revealed that this abnormal fiber pattern was associated with the anomalous course of the retinotectal fibers. No distinct differences in the cytoarchitecture and fiber pattern in the deep layers of the SC were seen. In conclusion, the present study demonstrated that the cytoarchitecture and fiber patterning in the superficial layers of the SC were disrupted in SRK rat, suggesting that Reelin protein regulates the formation of the superficial layers of the SC.


Subject(s)
Body Patterning/genetics , Cell Adhesion Molecules, Neuronal/deficiency , Extracellular Matrix Proteins/deficiency , Nerve Fibers, Myelinated/metabolism , Neurons/metabolism , Retina/abnormalities , Superior Colliculi/abnormalities , Visual Pathways/abnormalities , Animals , Cell Adhesion Molecules, Neuronal/genetics , Cell Communication/genetics , Cell Differentiation/genetics , Cholera Toxin/metabolism , Extracellular Matrix Proteins/genetics , Female , Fetus , Gene Deletion , Gene Expression Regulation, Developmental/genetics , Growth Cones/metabolism , Growth Cones/ultrastructure , Immunohistochemistry , Male , Nerve Fibers, Myelinated/ultrastructure , Nerve Tissue Proteins , Neurons/cytology , Pregnancy , Rats , Rats, Mutant Strains , Rats, Wistar , Reelin Protein , Retina/cytology , Retina/metabolism , Serine Endopeptidases , Superior Colliculi/cytology , Superior Colliculi/metabolism , Visual Pathways/cytology , Visual Pathways/metabolism
19.
Dev Cell ; 30(6): 673-87, 2014 Sep 29.
Article in English | MEDLINE | ID: mdl-25199687

ABSTRACT

In the process of neuronal wiring, axons derived from the same functional group typically extend together, resulting in fascicle formation. How these axons communicate with one another remains largely unknown. Here, we show that protocadherin-17 (Pcdh17) supports this group extension by recruiting actin polymerization regulators to interaxonal contact sites. Pcdh17 is expressed by a subset of amygdala neurons, and it accumulates at axon-axon boundaries because of homophilic binding. Pcdh17 knockout in mice suppressed the extension of these axons. Ectopically expressed Pcdh17 altered the pattern of axon extension. In in-vitro cultures, wild-type growth cones normally migrate along other axons, whereas Pcdh17 null growth cones do not. Pcdh17 recruits the WAVE complex, Lamellipodin, and Ena/VASP to cell-cell contacts, converting these sites into motile structures. We propose that, through these mechanisms, Pcdh17 maintains the migration of growth cones that are in contact with other axons, thereby supporting their collective extension.


Subject(s)
Actins/metabolism , Axons/metabolism , Cadherins/metabolism , Growth Cones/metabolism , Amygdala/cytology , Amygdala/growth & development , Amygdala/metabolism , Animals , Axons/physiology , Cadherins/genetics , Cell Movement , DNA-Binding Proteins/metabolism , Growth Cones/physiology , Mice , Protocadherins , Wiskott-Aldrich Syndrome Protein Family/metabolism
20.
Cell Rep ; 3(3): 919-30, 2013 Mar 28.
Article in English | MEDLINE | ID: mdl-23523355

ABSTRACT

Effective defense responses involve the entire organism. To maintain body homeostasis after tissue damage, a systemic wound response is induced in which the response of each tissue is tightly orchestrated to avoid incomplete recovery or an excessive, damaging response. Here, we provide evidence that in the systemic response to wounding, an apoptotic caspase pathway is activated downstream of reactive oxygen species in the midgut enterocytes (ECs), cells distant from the wound site, in Drosophila. We show that a caspase-pathway mutant has defects in homeostatic gut cell renewal and that inhibiting caspase activity in fly ECs results in the production of systemic lethal factors after wounding. Our results indicate that wounding remotely controls caspase activity in ECs, which activates the tissue stem cell regeneration pathway in the gut to dampen the dangerous systemic wound reaction.


Subject(s)
Apoptosis , Cell Proliferation , Drosophila/metabolism , Enterocytes/metabolism , Wound Healing , Animals , Caspases/genetics , Caspases/metabolism , Drosophila/physiology , Enterocytes/physiology , Homeostasis , Intestines/injuries , Mutation , Reactive Oxygen Species/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL