Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 69
Filter
Add more filters

Country/Region as subject
Publication year range
1.
J Med Genet ; 61(3): 239-243, 2024 Feb 21.
Article in English | MEDLINE | ID: mdl-37833059

ABSTRACT

DNA polymerase epsilon (Pol ε), a component of the core replisome, is involved in DNA replication. Although genetic defects of Pol ε have been reported to cause immunodeficiency syndromes, its role in haematopoiesis remains unknown. Here, we identified compound heterozygous variants (p.[Asp1131fs];[Thr1891del]) in POLE, encoding Pol ε catalytic subunit A (POLE1), in siblings with a syndromic form of severe congenital transfusion-dependent anaemia. In contrast to Diamond-Blackfan anaemia, marked reticulocytopenia or marked erythroid hypoplasia was not found. Their bone marrow aspirates during infancy revealed erythroid dysplasia with strongly positive TP53 in immunostaining. Repetitive examinations demonstrated trilineage myelodysplasia within 2 years from birth. They had short stature and facial dysmorphism. HEK293 cell-based expression experiments and analyses of patient-derived induced pluripotent stem cells (iPSCs) disclosed a reduced mRNA level of Asp1131fs-POLE1 and defective nuclear translocation of Thr1891del-POLE1. Analysis of iPSCs showed compensatory mRNA upregulation of the other replisome components and increase of the TP53 protein, both suggesting dysfunction of the replisome. We created Pole-knockout medaka fish and found that heterozygous fishes were viable, but with decreased RBCs. Our observations expand the phenotypic spectrum of the Pol ε defect in humans, additionally providing unique evidence linking Pol ε to haematopoiesis.


Subject(s)
DNA Polymerase II , DNA Replication , Animals , Humans , DNA Polymerase II/genetics , DNA Polymerase II/metabolism , HEK293 Cells , DNA Replication/genetics , Tumor Suppressor Protein p53/genetics , RNA, Messenger
2.
J Hum Genet ; 2024 Jun 20.
Article in English | MEDLINE | ID: mdl-38902431

ABSTRACT

Glycosylphosphatidylinositol (GPI)-anchored proteins are located at the cell surface by a covalent attachment between protein and GPI embedded in the plasma membrane. This attachment is catalyzed by GPI transamidase comprising five subunits (PIGK, PIGS, PIGT, PIGU, and GPAA1) in the endoplasmic reticulum. Loss of either subunit of GPI transamidase eliminates cell surface localization of GPI-anchored proteins. In humans, pathogenic variants in either subunit of GPI transamidase cause neurodevelopmental disorders. However, how the loss of GPI-anchored proteins triggers neurodevelopmental defects remains largely unclear. Here, we identified a novel homozygous variant of PIGK, NM_005482:c.481A > G,p. (Met161Val), in a Japanese female patient with neurodevelopmental delay, hypotonia, cerebellar atrophy, febrile seizures, hearing loss, growth impairment, dysmorphic facial features, and brachydactyly. The missense variant was found heterozygous in her father, but not in her mother. Zygosity analysis revealed that the homozygous PIGK variant in the patient was caused by paternal isodisomy. Rescue experiments using PIGK-deficient CHO cells revealed that the p.Met161Val variant of PIGK reduced GPI transamidase activity. Rescue experiments using pigk mutant zebrafish confirmed that the p.Met161Val variant compromised PIGK function in tactile-evoked motor response. We also demonstrated that axonal localization of voltage-gated sodium channels and concomitant generation of action potentials were impaired in pigk-deficient neurons in zebrafish, suggesting a link between GPI-anchored proteins and neuronal defects. Taken together, the missense p.Met161Val variant of PIGK is a novel pathogenic variant that causes the neurodevelopmental disorder.

3.
Dev Growth Differ ; 66(1): 43-55, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37779230

ABSTRACT

The freshwater planarian Dugesia japonica maintains an abundant heterogeneous cell population called neoblasts, which include adult pluripotent stem cells. Thus, it is an excellent model organism for stem cell and regeneration research. Recently, many single-cell RNA sequencing (scRNA-seq) databases of several model organisms, including other planarian species, have become publicly available; these are powerful and useful resources to search for gene expression in various tissues and cells. However, the only scRNA-seq dataset for D. japonica has been limited by the number of genes detected. Herein, we collected D. japonica cells, and conducted an scRNA-seq analysis. A novel, automatic, iterative cell clustering strategy produced a dataset of 3,404 cells, which could be classified into 63 cell types based on gene expression profiles. We introduced two examples for utilizing the scRNA-seq dataset in this study using D. japonica. First, the dataset provided results consistent with previous studies as well as novel functionally relevant insights, that is, the expression of DjMTA and DjP2X-A genes in neoblasts that give rise to differentiated cells. Second, we conducted an integrative analysis of the scRNA-seq dataset and time-course bulk RNA-seq of irradiated animals, demonstrating that the dataset can help interpret differentially expressed genes captured via bulk RNA-seq. Using the R package "Seurat" and GSE223927, researchers can easily access and utilize this dataset.


Subject(s)
Adult Stem Cells , Planarians , Pluripotent Stem Cells , Animals , Planarians/genetics , Planarians/metabolism , Transcriptome/genetics , Gene Expression Profiling
4.
Mov Disord ; 37(10): 2139-2146, 2022 10.
Article in English | MEDLINE | ID: mdl-35876425

ABSTRACT

BACKGROUND: Despite advances in next generation sequencing technologies, the identification of variants of uncertain significance (VUS) can often hinder definitive diagnosis in patients with complex neurodevelopmental disorders. OBJECTIVE: The objective of this study was to identify and characterize the underlying cause of disease in a family with two children with severe developmental delay associated with generalized dystonia and episodic status dystonicus, chorea, epilepsy, and cataracts. METHODS: Candidate genes identified by autozygosity mapping and whole-exome sequencing were characterized using cellular and vertebrate model systems. RESULTS: Homozygous variants were found in three candidate genes: MED27, SLC6A7, and MPPE1. Although the patients had features of MED27-related disorder, the SLC6A7 and MPPE1 variants were functionally investigated. SLC6A7 variant in vitro overexpression caused decreased proline transport as a result of reduced cell-surface expression, and zebrafish knockdown of slc6a7 exhibited developmental delay and fragile motor neuron morphology that could not be rescued by L-proline transporter-G396S RNA. Lastly, patient fibroblasts displayed reduced cell-surface expression of glycophosphatidylinositol-anchored proteins linked to MPPE1 dysfunction. CONCLUSIONS: We report a family harboring a homozygous MED27 variant with additional loss-of-function SLC6A7 and MPPE1 gene variants, which potentially contribute to a blended phenotype caused by multilocus pathogenic variants. © 2022 The Authors. Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society.


Subject(s)
Dystonia , Dystonic Disorders , Movement Disorders , Neurodevelopmental Disorders , Animals , Dystonia/diagnosis , Dystonia/genetics , Dystonic Disorders/genetics , Movement Disorders/genetics , Neurodevelopmental Disorders/genetics , Proline , RNA , Zebrafish/genetics
5.
Am J Med Genet A ; 188(4): 1184-1192, 2022 04.
Article in English | MEDLINE | ID: mdl-35018717

ABSTRACT

Nuclear factor one A (NFIA) is a transcription factor that regulates the development of the central nervous system. Haploinsufficiency of the NFIA gene causes NFIA-related disorder, which includes brain abnormalities and intellectual disability, with or without urinary tract defects. Intragenic deletions, nonsense variants, frameshift variants, and missense variants in one allele of the NFIA gene have been reported to cause various neurological and urogenital symptoms. Here we report a 10-year-old male patient with developmental delay, coarctation of the aorta, and distinctive facial features. Exome analysis identified a rare de novo heterozygous missense variant p.Thr395Met in NFIA. We employed zebrafish as a model organism in our NFIA analysis and found that nfia-/- zebrafish initially showed a loss of commissural axons in the brain, and eventually underwent growth retardation resulting in premature death. Impairment of the commissural neurons in nfia-/- zebrafish embryos could be restored by the expression of wild-type human NFIA protein, but not of mutant human protein harboring the p.Thr395Met substitution, indicating that this variant affects the function of NFIA protein. Taken together, we suggest that the p.Thr395Met allele in the NFIA gene is relevant to the pathogenesis of NFIA-related disorder.


Subject(s)
Intellectual Disability , Neurodevelopmental Disorders , Animals , Haploinsufficiency , Humans , Intellectual Disability/genetics , Intellectual Disability/pathology , Male , Mutation, Missense/genetics , NFI Transcription Factors/genetics , Neurodevelopmental Disorders/genetics , Zebrafish/genetics
6.
Ann Neurol ; 88(3): 544-561, 2020 09.
Article in English | MEDLINE | ID: mdl-32588476

ABSTRACT

OBJECTIVE: Impairment of glycinergic neurotransmission leads to complex movement and behavioral disorders. Patients harboring glycine receptor autoantibodies suffer from stiff-person syndrome or its severe variant progressive encephalomyelitis with rigidity and myoclonus. Enhanced receptor internalization was proposed as the common molecular mechanism upon autoantibody binding. Although functional impairment of glycine receptors following autoantibody binding has recently been investigated, it is still incompletely understood. METHODS: A cell-based assay was used for positive sample evaluation. Glycine receptor function was assessed by electrophysiological recordings and radioligand binding assays. The in vivo passive transfer of patient autoantibodies was done using the zebrafish animal model. RESULTS: Glycine receptor function as assessed by glycine dose-response curves showed significantly decreased glycine potency in the presence of patient sera. Upon binding of autoantibodies from 2 patients, a decreased fraction of desensitized receptors was observed, whereas closing of the ion channel remained fast. The glycine receptor N-terminal residues 29 A to 62 G were mapped as a common epitope of glycine receptor autoantibodies. An in vivo transfer into the zebrafish animal model generated a phenotype with disturbed escape behavior accompanied by a reduced number of glycine receptor clusters in the spinal cord of affected animals. INTERPRETATION: Autoantibodies against the extracellular domain mediate alterations of glycine receptor physiology. Moreover, our in vivo data demonstrate that the autoantibodies are a direct cause of the disease, because the transfer of human glycine receptor autoantibodies to zebrafish larvae generated impaired escape behavior in the animal model compatible with abnormal startle response in stiff-person syndrome or progressive encephalitis with rigidity and myoclonus patients. ANN NEUROL 2020;88:544-561.


Subject(s)
Autoantibodies/immunology , Encephalomyelitis/immunology , Muscle Rigidity/immunology , Receptors, Glycine/metabolism , Stiff-Person Syndrome/immunology , Adult , Aged , Animals , Autoantibodies/pharmacology , Autoantigens/immunology , Behavior, Animal/drug effects , Encephalomyelitis/metabolism , Epitopes, B-Lymphocyte/immunology , Female , Humans , Male , Middle Aged , Muscle Rigidity/metabolism , Receptors, Glycine/immunology , Stiff-Person Syndrome/metabolism , Zebrafish
7.
Am J Med Genet A ; 185(7): 2084-2093, 2021 07.
Article in English | MEDLINE | ID: mdl-33973697

ABSTRACT

Nuclear factor I A (NFIA) is a transcription factor that belongs to the NFI family. Truncating variants or intragenic deletion of the NFIA gene are known to cause the human neurodevelopmental disorder known as NFIA-related disorder, but no patient heterozygous for a missense mutation has been reported. Here, we document two unrelated patients with typical phenotypic features of the NFIA-related disorder who shared a missense variant p.Lys125Glu (K125E) in the NFIA gene. Patient 1 was a 6-year-old female with global developmental delay, corpus callosum anomaly, macrocephaly, and dysmorphic facial features. Patient 2 was a 14-month-old male with corpus callosum anomaly and macrocephaly. By using Drosophila and zebrafish models, we functionally evaluated the effect of the K125E substitution. Ectopic expression of wild-type human NFIA in Drosophila caused developmental defects such as eye malformation and premature death, while that of human NFIA K125E variant allele did not. nfia-deficient zebrafish embryos showed defects of midline-crossing axons in the midbrain/hindbrain boundary. This impairment of commissural neurons was rescued by expression of wild-type human NFIA, but not by that of mutant variant harboring K125E substitution. In accordance with these in vivo functional analyses, we showed that the K125E mutation impaired the transcriptional regulation of HES1 promoter in cultured cells. Taken together, we concluded that the K125E variant in the NFIA gene is a loss-of-function mutation.


Subject(s)
Genetic Predisposition to Disease , Megalencephaly/genetics , NFI Transcription Factors/genetics , Neurodevelopmental Disorders/genetics , Alleles , Amino Acid Substitution/genetics , Animals , Child , Corpus Callosum/metabolism , Corpus Callosum/pathology , Disease Models, Animal , Drosophila/genetics , Female , Gene Expression Regulation, Developmental/genetics , Humans , Infant , Male , Megalencephaly/pathology , Mutation, Missense/genetics , Neurodevelopmental Disorders/pathology , Zebrafish/genetics
8.
J Neurosci ; 39(45): 8988-8997, 2019 11 06.
Article in English | MEDLINE | ID: mdl-31558619

ABSTRACT

The process by which future behavioral responses are shaped by past experiences is one of the central questions in neuroscience. To gain insight into this process at the molecular and cellular levels, we have applied zebrafish larvae to explore behavioral desensitization to sound. A sudden loud noise often evokes a defensive response known as the acoustic startle response (ASR), which is triggered by firing Mauthner cells in teleosts and amphibians. The probability of evoking ASR by suprathreshold sound is reduced after exposure to repetitive auditory stimuli insufficient in amplitude to evoke the ASR (subthreshold). Although it has been suggested that the potentiation of inhibitory glycinergic inputs into Mauthner cell is involved in this desensitization of the ASR, the molecular basis for the potentiation of glycinergic transmission has been unclear. Through the in vivo monitoring of fluorescently-tagged glycine receptors (GlyRs), we here showed that behavioral desensitization to sound in zebrafish is governed by GlyR clustering in Mauthner cells. We further revealed that CaMKII-dependent phosphorylation of the scaffolding protein gephyrin at serine 325 promoted the synaptic accumulation of GlyR on Mauthner neurons through the enhancement of the gephyrin-GlyR binding, which was indispensable for and could induce desensitization of the ASR. Our study demonstrates an essential molecular and cellular basis of sound-induced receptor dynamics and thus of behavioral desensitization to sound.SIGNIFICANCE STATEMENT Behavioral desensitization in the acoustic startle response of fish is known to involve the potentiation of inhibitory glycinergic input to the Mauthner cell, which is a command neuron for the acoustic startle response. However, the molecular and cellular basis for this potentiation has been unknown. Here we show that an increase in glycine receptor (GlyR) clustering at synaptic sites on zebrafish Mauthner cells is indispensable for and could induce desensitization. Furthermore, we demonstrate that CaMKII-mediated phosphorylation of the scaffolding protein gephyrin promotes GlyR clustering by increasing the binding between the ß-loop of GlyRs and gephyrin. Thus, the phosphorylation of gephyrin is a key event which accounts for the potentiation of inhibitory glycinergic inputs observed during sound-evoked behavioral desensitization.


Subject(s)
Auditory Perception , Membrane Proteins/metabolism , Neurons/metabolism , Receptors, Glycine/metabolism , Reflex, Startle , Zebrafish Proteins/metabolism , Animals , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Neurons/physiology , Phosphorylation , Synapses/metabolism , Synapses/physiology , Zebrafish
9.
Hum Mutat ; 40(12): 2270-2285, 2019 12.
Article in English | MEDLINE | ID: mdl-31206972

ABSTRACT

Pathogenic variants in the X-linked gene ZC4H2, which encodes a zinc-finger protein, cause an infrequently described syndromic form of arthrogryposis multiplex congenita (AMC) with central and peripheral nervous system involvement. We present genetic and detailed phenotypic information on 23 newly identified families and simplex cases that include 19 affected females from 18 families and 14 affected males from nine families. Of note, the 15 females with deleterious de novo ZC4H2 variants presented with phenotypes ranging from mild to severe, and their clinical features overlapped with those seen in affected males. By contrast, of the nine carrier females with inherited ZC4H2 missense variants that were deleterious in affected male relatives, four were symptomatic. We also compared clinical phenotypes with previously published cases of both sexes and provide an overview on 48 males and 57 females from 42 families. The spectrum of ZC4H2 defects comprises novel and recurrent mostly inherited missense variants in affected males, and de novo splicing, frameshift, nonsense, and partial ZC4H2 deletions in affected females. Pathogenicity of two newly identified missense variants was further supported by studies in zebrafish. We propose ZC4H2 as a good candidate for early genetic testing of males and females with a clinical suspicion of fetal hypo-/akinesia and/or (neurogenic) AMC.


Subject(s)
Arthrogryposis/genetics , Intracellular Signaling Peptides and Proteins/genetics , Mutation , Nuclear Proteins/genetics , Animals , Codon, Nonsense , Disease Models, Animal , Female , Frameshift Mutation , Genes, X-Linked , Genetic Predisposition to Disease , Humans , Male , Mutation, Missense , Pedigree , Phenotype , Sequence Deletion , Sex Characteristics , Zebrafish
10.
Am J Hum Genet ; 98(3): 473-489, 2016 Mar 03.
Article in English | MEDLINE | ID: mdl-26924529

ABSTRACT

Transcriptional signal cointegrators associate with transcription factors or nuclear receptors and coregulate tissue-specific gene transcription. We report on recessive loss-of-function mutations in two genes (TRIP4 and ASCC1) that encode subunits of the nuclear activating signal cointegrator 1 (ASC-1) complex. We used autozygosity mapping and whole-exome sequencing to search for pathogenic mutations in four families. Affected individuals presented with prenatal-onset spinal muscular atrophy (SMA), multiple congenital contractures (arthrogryposis multiplex congenita), respiratory distress, and congenital bone fractures. We identified homozygous and compound-heterozygous nonsense and frameshift TRIP4 and ASCC1 mutations that led to a truncation or the entire absence of the respective proteins and cosegregated with the disease phenotype. Trip4 and Ascc1 have identical expression patterns in 17.5-day-old mouse embryos with high expression levels in the spinal cord, brain, paraspinal ganglia, thyroid, and submandibular glands. Antisense morpholino-mediated knockdown of either trip4 or ascc1 in zebrafish disrupted the highly patterned and coordinated process of α-motoneuron outgrowth and formation of myotomes and neuromuscular junctions and led to a swimming defect in the larvae. Immunoprecipitation of the ASC-1 complex consistently copurified cysteine and glycine rich protein 1 (CSRP1), a transcriptional cofactor, which is known to be involved in spinal cord regeneration upon injury in adult zebrafish. ASCC1 mutant fibroblasts downregulated genes associated with neurogenesis, neuronal migration, and pathfinding (SERPINF1, DAB1, SEMA3D, SEMA3A), as well as with bone development (TNFRSF11B, RASSF2, STC1). Our findings indicate that the dysfunction of a transcriptional coactivator complex can result in a clinical syndrome affecting the neuromuscular system.


Subject(s)
Fractures, Bone/genetics , Gene Expression Regulation, Developmental , Muscular Atrophy, Spinal/genetics , Transcription Factors/genetics , Amino Acid Sequence , Animals , Arthrogryposis/diagnosis , Arthrogryposis/genetics , Carrier Proteins/genetics , Cells, Cultured , Fibroblasts/cytology , Fibroblasts/metabolism , Fractures, Bone/diagnosis , Gene Expression Profiling , Homozygote , Humans , LIM Domain Proteins/genetics , Mice , Molecular Sequence Data , Muscular Atrophy, Spinal/diagnosis , Mutation , Nuclear Proteins/genetics , Pedigree , Phenotype , Zebrafish , Zebrafish Proteins/genetics
11.
Genes Cells ; 23(11): 938-951, 2018 Nov.
Article in English | MEDLINE | ID: mdl-30151851

ABSTRACT

Integrins are transmembrane molecules that facilitate cell-to-cell and cell-to-extracellular matrix (ECM) interactions. Integrin molecules are heterodimers that consist of α- and ß-subunits. The integrin ß1 gene is widely expressed in vivo and is the major ß molecule in many tissues; however, tissue-specific roles of integrin ß1 are still elusive. In this study, we investigated integrin ß1 function in endothelial cells of zebrafish. An integrin ß1b mutant zebrafish exhibited morphological abnormalities in blood vessel formation, cephalic hemorrhage and a decreased responsiveness to tactile stimulation during development. To determine the role of integrin ß1b in vascular formation, we developed a Gal4/UAS-mediated conditional inactivation of integrin ß1 by expressing the cytoplasmic region of integrin ß1 that acts as a dominant-negative (DN) isoform. Expression of integrin ß1 DN in endothelial cells induced blood vessel abnormalities as in integrin ß1b mutants. These results show that endothelial cells require integrin activity for the formation and/or maintenance of blood vessels in zebrafish. Furthermore, our time-lapse recording visualized the breakpoint of cephalic vessels and the hemorrhage onset. Taken together, our tissue-specific inactivation of integrin ß1 in zebrafish is powerful tools for functional analysis of integrin ß1 in developing tissues.


Subject(s)
Cardiovascular Abnormalities/pathology , Embryo, Nonmammalian/pathology , Endothelium, Vascular/pathology , Hemorrhage/pathology , Integrin beta1/metabolism , Mutation , Zebrafish/embryology , Animals , Animals, Genetically Modified/embryology , Animals, Genetically Modified/genetics , Animals, Genetically Modified/metabolism , Cardiovascular Abnormalities/genetics , Cardiovascular Abnormalities/metabolism , Embryo, Nonmammalian/metabolism , Hemorrhage/genetics , Hemorrhage/metabolism , Integrin beta1/genetics , Zebrafish/genetics , Zebrafish/metabolism
12.
Proc Natl Acad Sci U S A ; 112(9): 2859-64, 2015 Mar 03.
Article in English | MEDLINE | ID: mdl-25691753

ABSTRACT

Following their synthesis in the endoplasmic reticulum (ER), voltage-gated sodium channels (NaV) are transported to the membranes of excitable cells, where they often cluster, such as at the axon initial segment of neurons. Although the mechanisms by which NaV channels form and maintain clusters have been extensively examined, the processes that govern their transport and degradation have received less attention. Our entry into the study of these processes began with the isolation of a new allele of the zebrafish mutant alligator, which we found to be caused by mutations in the gene encoding really interesting new gene (RING) finger protein 121 (RNF121), an E3-ubiquitin ligase present in the ER and cis-Golgi compartments. Here we demonstrate that RNF121 facilitates two opposing fates of NaV channels: (i) ubiquitin-mediated proteasome degradation and (ii) membrane localization when coexpressed with auxiliary NaVß subunits. Collectively, these results indicate that RNF121 participates in the quality control of NaV channels during their synthesis and subsequent transport to the membrane.


Subject(s)
Proteolysis , RING Finger Domains , Ubiquitin-Protein Ligases/metabolism , Voltage-Gated Sodium Channels/metabolism , Zebrafish Proteins/metabolism , Zebrafish/metabolism , Animals , Base Sequence , Cell Membrane/genetics , Cell Membrane/metabolism , Molecular Sequence Data , Mutation , Proteasome Endopeptidase Complex/genetics , Proteasome Endopeptidase Complex/metabolism , Protein Transport/physiology , Ubiquitin/genetics , Ubiquitin/metabolism , Ubiquitin-Protein Ligases/genetics , Voltage-Gated Sodium Channels/genetics , Zebrafish/genetics , Zebrafish Proteins/genetics
13.
J Biol Chem ; 291(47): 24465-24474, 2016 Nov 18.
Article in English | MEDLINE | ID: mdl-27681595

ABSTRACT

In the retina, aberrant opsin transport from cell bodies to outer segments leads to retinal degenerative diseases such as retinitis pigmentosa. Opsin transport is facilitated by the intraflagellar transport (IFT) system that mediates the bidirectional movement of proteins within cilia. In contrast to functions of the anterograde transport executed by IFT complex B (IFT-B), the precise functions of the retrograde transport mediated by IFT complex A (IFT-A) have not been well studied in photoreceptor cilia. Here, we analyzed developing zebrafish larvae carrying a null mutation in ift122 encoding a component of IFT-A. ift122 mutant larvae show unexpectedly mild phenotypes, compared with those of mutants defective in IFT-B. ift122 mutants exhibit a slow onset of progressive photoreceptor degeneration mainly after 7 days post-fertilization. ift122 mutant larvae also develop cystic kidney but not curly body, both of which are typically observed in various ciliary mutants. ift122 mutants display a loss of cilia in the inner ear hair cells and nasal pit epithelia. Loss of ift122 causes disorganization of outer segment discs. Ectopic accumulation of an IFT-B component, ift88, is observed in the ift122 mutant photoreceptor cilia. In addition, pulse-chase experiments using GFP-opsin fusion proteins revealed that ift122 is required for the efficient transport of opsin and the distal elongation of outer segments. These results show that IFT-A is essential for the efficient transport of outer segment proteins, including opsin, and for the survival of retinal photoreceptor cells, rendering the ift122 mutant a unique model for human retinal degenerative diseases.


Subject(s)
Opsins/metabolism , Photoreceptor Cells, Vertebrate/metabolism , Retinal Degeneration/metabolism , Zebrafish Proteins/metabolism , Zebrafish/metabolism , Animals , Cilia/genetics , Cilia/metabolism , Hair Cells, Auditory, Inner/metabolism , Humans , Mutation , Opsins/genetics , Protein Transport/genetics , Retinal Degeneration/genetics , Zebrafish/genetics , Zebrafish Proteins/genetics
14.
Am J Hum Genet ; 92(5): 681-95, 2013 May 02.
Article in English | MEDLINE | ID: mdl-23623388

ABSTRACT

Arthrogryposis multiplex congenita (AMC) is caused by heterogeneous pathologies leading to multiple antenatal joint contractures through fetal akinesia. Understanding the pathophysiology of this disorder is important for clinical care of the affected individuals and genetic counseling of the families. We thus aimed to establish the genetic basis of an AMC subtype that is associated with multiple dysmorphic features and intellectual disability (ID). We used haplotype analysis, next-generation sequencing, array comparative genomic hybridization, and chromosome breakpoint mapping to identify the pathogenic mutations in families and simplex cases. Suspected disease variants were verified by cosegregation analysis. We identified disease-causing mutations in the zinc-finger gene ZC4H2 in four families affected by X-linked AMC plus ID and one family affected by cerebral palsy. Several heterozygous females were also affected, but to a lesser degree. Furthermore, we found two ZC4H2 deletions and one rearrangement in two female and one male unrelated simplex cases, respectively. In mouse primary hippocampal neurons, transiently produced ZC4H2 localized to the postsynaptic compartment of excitatory synapses, and the altered protein influenced dendritic spine density. In zebrafish, antisense-morpholino-mediated zc4h2 knockdown caused abnormal swimming and impaired α-motoneuron development. All missense mutations identified herein failed to rescue the swimming defect of zebrafish morphants. We conclude that ZC4H2 point mutations, rearrangements, and small deletions cause a clinically variable broad-spectrum neurodevelopmental disorder of the central and peripheral nervous systems in both familial and simplex cases of both sexes. Our results highlight the importance of ZC4H2 for genetic testing of individuals presenting with ID plus muscle weakness and minor or major forms of AMC.


Subject(s)
Abnormalities, Multiple/genetics , Arthrogryposis/genetics , Carrier Proteins/genetics , Genetic Predisposition to Disease/genetics , Intellectual Disability/genetics , Neuronal Plasticity/genetics , Zinc Fingers/genetics , Abnormalities, Multiple/pathology , Animals , Arthrogryposis/pathology , Cells, Cultured , Chromosome Breakpoints , Comparative Genomic Hybridization , Female , Haplotypes/genetics , High-Throughput Nucleotide Sequencing , Humans , Immunoblotting , In Situ Hybridization , Intellectual Disability/pathology , Intracellular Signaling Peptides and Proteins , Male , Mice , Mutation/genetics , Nuclear Proteins , Pedigree , Synapses/genetics , Zebrafish
15.
Development ; 140(18): 3927-31, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23946442

ABSTRACT

The developing nervous system consists of a variety of cell types. Transgenic animals expressing reporter genes in specific classes of neuronal cells are powerful tools for the study of neuronal network formation. We generated a wide variety of transgenic zebrafish that expressed reporter genes in specific classes of neurons or neuronal progenitors. These include lines in which neurons of specific neurotransmitter phenotypes expressed fluorescent proteins or Gal4, and lines in which specific subsets of the dorsal progenitor domain in the spinal cord expressed fluorescent proteins. Using these, we examined domain organization in the developing dorsal spinal cord, and found that there are six progenitor domains in zebrafish, which is similar to the domain organization in mice. We also systematically characterized neurotransmitter properties of the neurons that are produced from each domain. Given that reporter gene expressions occurs in a wide area of the nervous system in the lines generated, these transgenic fish should serve as powerful tools for the investigation of not only the neurons in the dorsal spinal cord but also neuronal structures and functions in many other regions of the nervous system.


Subject(s)
Gene Transfer Techniques , Neurons/cytology , Neurons/metabolism , Zebrafish/genetics , Animals , Animals, Genetically Modified , Mice , Neurotransmitter Agents/metabolism , Phenotype , Spinal Cord/cytology , Spinal Cord/metabolism
16.
J Gerontol Nurs ; 41(4): 38-46, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25347865

ABSTRACT

Aggressive behaviors (ABs) related to dementia in older adults have been associated with increased occupational stress among care workers (CWs) in the United States and other Western countries, and they may contribute to staff turnover. However, few studies related to this issue have been conducted in Japan. The current cross-sectional study examined (a) the relationship between CW frequency of exposure to dementia-related ABs and CW occupational stress (i.e., job burnout, job satisfaction, and intention to resign), and (b) mediator effects between frequency of exposure to dementia-related ABs and CW occupational stress. A total of 137 CWs in dementia special care units from 10 nursing homes in Japan were recruited as study participants. Major findings indicate that the relationship between exposure to ABs and work outcomes was fully mediated by the appraisal of stress. Findings from this study may be used to develop culturally relevant training and educational interventions targeted at reducing ABs in individuals with dementia and occupational stress from exposure to ABs among CWs.


Subject(s)
Aggression , Burnout, Professional/epidemiology , Dementia/nursing , Homes for the Aged/statistics & numerical data , Nursing Homes/statistics & numerical data , Occupational Exposure/statistics & numerical data , Workplace Violence/statistics & numerical data , Adult , Aged , Aged, 80 and over , Burnout, Professional/psychology , Cross-Sectional Studies , Dementia/epidemiology , Environmental Monitoring , Female , Health Personnel/psychology , Health Personnel/statistics & numerical data , Humans , Incidence , Japan/epidemiology , Job Satisfaction , Male , Middle Aged , Personnel Turnover/statistics & numerical data , Stress, Psychological/epidemiology , Workplace Violence/psychology
17.
J Neurosci ; 33(37): 14638-44, 2013 Sep 11.
Article in English | MEDLINE | ID: mdl-24027265

ABSTRACT

RNA helicases regulate RNA metabolism, but their substrate specificity and in vivo function remain largely unknown. We isolated spontaneous mutant zebrafish that exhibit an abnormal dorsal bend at the beginning of tactile-evoked escape swimming. Similar behavioral defects were observed in zebrafish embryos treated with strychnine, which blocks glycine receptors (GlyRs), suggesting that the abnormal motor response in mutants may be attributable to a deficit in glycinergic synaptic transmission. We identified a missense mutation in the gene encoding RNA helicase Dhx37. In Dhx37 mutants, ribosomal RNA levels were unchanged, whereas GlyR α1, α3, and α4a subunit mRNA levels were decreased due to a splicing defect. We found that Dhx37 can interact with GlyR α1, α3, and α4a transcripts but not with the GlyR α2 subunit mRNA. Overexpression of GlyR α1, α3, or α4a subunits in Dhx37-deficient embryos restored normal behavior. Conversely, antisense-mediated knockdown of multiple GlyR α subunits in wild-type embryos was required to recapitulate the Dhx37 mutant phenotype. These results indicate that Dhx37 is specifically required for the biogenesis of a subset of GlyR α subunit mRNAs, thereby regulating glycinergic synaptic transmission and associated motor behaviors. To our knowledge, this is the first identification of pathologically relevant substrates for an RNA helicase.


Subject(s)
DEAD-box RNA Helicases/genetics , Escape Reaction/physiology , Gene Expression Regulation, Developmental/genetics , Mutation/genetics , Receptors, Glycine/metabolism , Animals , Animals, Genetically Modified , Brain/cytology , Embryo, Nonmammalian , Gene Expression Regulation, Developmental/drug effects , Glycine Agents/pharmacology , Mutation, Missense , Oligodeoxyribonucleotides, Antisense/pharmacology , Patch-Clamp Techniques , Physical Stimulation/adverse effects , Protein Subunits/genetics , Protein Subunits/metabolism , RNA, Messenger/metabolism , Receptors, Glycine/genetics , Strychnine/pharmacology , Swimming/physiology , Synapses/drug effects , Synaptic Transmission/drug effects , Synaptic Transmission/genetics , Video Recording , Zebrafish
18.
J Neurophysiol ; 111(6): 1153-64, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24335214

ABSTRACT

Each neuron possesses a unique firing property, which is largely attributed to heterogeneity in the composition of voltage-gated ion channel complexes. Zebrafish Mauthner (M) cells, which are bilaterally paired giant reticulospinal neurons (RSNs) in the hindbrain and induce rapid escape behavior, generate only a single spike at the onset of depolarization. This single spiking is in contrast with the repetitive firing of the M cell's morphologically homologous RSNs, MiD2cm and MiD3cm, which are also involved in escapes. However, how the unique firing property of M cells is established and the underlying molecular mechanisms remain unclear. In the present study, we first demonstrated that the single-spiking property of M cells was acquired at 4 days postfertilization (dpf), accompanied by an increase in dendrotoxin I (DTX)-sensitive low-threshold K(+) currents, prior to which the M cell repetitively fires as its homologs. Second, in situ hybridization showed that among DTX-sensitive Kv1 channel α-subunits, zKv1.1a was unexpectedly expressed even in the homologs and the bursting M cells at 2 dpf. In contrast, zKvß2b, an auxiliary ß-subunit of Kv1 channels, was expressed only in the single-spiking M cells. Third, zKv1.1a expressed in Xenopus oocytes functioned as a low-threshold K(+) channel, and its currents were enhanced by coexpression of zKvß2b subunits. Finally, knockdown of zKvß2b expression in zebrafish larvae resulted in repetitive firing of M cells at 4 dpf. Taken together, these results suggest that associative expression of Kvß2 subunits with Kv1.1 channels is crucial for developmental acquisition of the unique firing properties of the M cells among homologous neurons.


Subject(s)
Action Potentials , Kv1.1 Potassium Channel/metabolism , Neurons/physiology , Zebrafish Proteins/metabolism , Animals , Elapid Venoms/pharmacology , Kv1.1 Potassium Channel/antagonists & inhibitors , Kv1.1 Potassium Channel/genetics , Neurons/metabolism , Potassium Channel Blockers/pharmacology , Protein Multimerization , Protein Subunits/antagonists & inhibitors , Protein Subunits/genetics , Protein Subunits/metabolism , Rhombencephalon/cytology , Rhombencephalon/growth & development , Rhombencephalon/physiology , Zebrafish , Zebrafish Proteins/antagonists & inhibitors , Zebrafish Proteins/genetics
19.
Genes Cells ; 18(3): 211-24, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23347046

ABSTRACT

Synaptic transmission-dependent regulation of neurotransmitter receptor accumulation at postsynaptic sites underlies the formation, maintenance and maturation of synaptic function. Previous in vitro studies showed that glycine receptor (GlyR) clustering requires synaptic inputs. However, in vivo GlyR regulation by synaptic transmission is not fully understood. Here, we established a model system using developing zebrafish, in which GlyRs are expressed in Mauthner cells (M-cells), a pair of giant, reticulospinal, hindbrain neurons, thereby enabling analysis of GlyR clusters over time in identifiable cells. Bath application of a glycinergic blocker, strychnine, to developing zebrafish prevented postsynaptic GlyR cluster formation in the M-cells. After strychnine removal, the GlyR clusters appeared in the M-cells. At a later stage, glycinergic transmission blockade impaired maintenance of GlyR clusters. We also found that pharmacological blockade of either L-type Ca(2+) channels or calcium-/calmodulin-dependent protein kinase II (CaMKII) disturbed GlyR clustering. In addition, the M-cell-specific CaMKII inactivation using the Gal4-UAS system significantly impaired GlyR clustering in the M-cells. Thus, the formation and maintenance of GlyR clusters in the M-cells in the developing animals are regulated in a synaptic transmission-dependent manner, and CaMKII activation at the postsynapse is essential for GlyR clustering. This is the first demonstration of synaptic transmission-dependent modulation of synaptic GlyRs in vivo.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Glycine/metabolism , Receptors, Glycine/metabolism , Synaptic Transmission , Animals , Calcium Channel Blockers/pharmacology , Calcium Channels, L-Type/drug effects , Calcium Channels, L-Type/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Type 2/antagonists & inhibitors , Neurons/metabolism , Receptors, Glycine/antagonists & inhibitors , Rhombencephalon/cytology , Rhombencephalon/growth & development , Strychnine/pharmacology , Synapses/drug effects , Synapses/metabolism , Synapses/ultrastructure , Synaptic Transmission/drug effects , Zebrafish/metabolism
20.
Sci Rep ; 14(1): 7455, 2024 03 29.
Article in English | MEDLINE | ID: mdl-38548817

ABSTRACT

Inbred strains of organisms are genetically highly uniform and thus useful for life science research. We have previously reported the ongoing generation of the zebrafish IM strain from the India (IND) strain through full sib-pair mating for 16 generations. However, the IM fish laid a small number of offspring and had a short lifespan, implying the need for discreet care in breeding. Here, we report the subsequent establishment of IM strain as well as the generation of a new inbred zebrafish strain, Mishima-AB (M-AB). M-AB was derived from the *AB strain by full sib-pair mating for over 20 generations, which fulfills the general criterion for the establishment of an inbred strain. In contrast to the IM case, maintenance of the M-AB strain by sib-pair mating required almost no special handling. Genome sequencing of IM individuals from the 47th generation and M-AB individuals from the 27th generation revealed that SNP-based genomic heterogeneity across whole-genome nucleotides was 0.008% and 0.011%, respectively. These percentages were much lower than those of the parental IND (0.197%) and *AB (0.086%) strains. These results indicate that the genomes of these inbred strains were highly homogenous. We also demonstrated the successful microinjection of antisense morpholinos, CRISPR/Cas9, and foreign genes into M-AB embryos at the 1-cell stage. Overall, we report the establishment of a zebrafish inbred strain, M-AB, which is capable of regular breeding and genetic manipulation. This strain will be useful for the analysis of genetically susceptible phenotypes such as behaviors, microbiome features and drug susceptibility.


Subject(s)
Inbreeding , Zebrafish , Animals , Zebrafish/genetics , Genome , Chromosome Mapping , Phenotype
SELECTION OF CITATIONS
SEARCH DETAIL