Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 51
Filter
Add more filters

Country/Region as subject
Publication year range
1.
J Biol Chem ; 295(27): 9244-9262, 2020 07 03.
Article in English | MEDLINE | ID: mdl-32434929

ABSTRACT

Calsyntenin-3 (Clstn3) is a postsynaptic adhesion molecule that induces presynaptic differentiation via presynaptic neurexins (Nrxns), but whether Nrxns directly bind to Clstn3 has been a matter of debate. Here, using LC-MS/MS-based protein analysis, confocal microscopy, RNAscope assays, and electrophysiological recordings, we show that ß-Nrxns directly interact via their LNS domain with Clstn3 and Clstn3 cadherin domains. Expression of splice site 4 (SS4) insert-positive ß-Nrxn variants, but not insert-negative variants, reversed the impaired Clstn3 synaptogenic activity observed in Nrxn-deficient neurons. Consistently, Clstn3 selectively formed complexes with SS4-positive Nrxns in vivo Neuron-specific Clstn3 deletion caused significant reductions in number of excitatory synaptic inputs. Moreover, expression of Clstn3 cadherin domains in CA1 neurons of Clstn3 conditional knockout mice rescued structural deficits in excitatory synapses, especially within the stratum radiatum layer. Collectively, our results suggest that Clstn3 links to SS4-positive Nrxns to induce presynaptic differentiation and orchestrate excitatory synapse development in specific hippocampal neural circuits, including Schaffer collateral afferents.


Subject(s)
Calcium-Binding Proteins/metabolism , Membrane Proteins/metabolism , Nerve Tissue Proteins/metabolism , Neural Cell Adhesion Molecules/metabolism , Animals , Cadherins/metabolism , Calcium-Binding Proteins/physiology , Chromatography, Liquid/methods , Hippocampus/metabolism , Membrane Proteins/physiology , Mice , Nerve Tissue Proteins/physiology , Neural Cell Adhesion Molecules/physiology , Neurons/metabolism , Synapses/metabolism , Tandem Mass Spectrometry/methods
2.
Am J Med Genet A ; 185(10): 3092-3098, 2021 10.
Article in English | MEDLINE | ID: mdl-34042275

ABSTRACT

Angelman syndrome is a neurodevelopmental disorder characterized by intellectual disability (ID), a distinctive gait pattern, abnormal behaviors, severe impairment in language development, and characteristic facial features. Most cases are caused by the absence of a maternal contribution to the imprinted region on chromosome 15q11-q13. Here, we present the first reported case of a 3-year-old boy with an atypical phenotype of Angelman syndrome due to uniparental isodisomy with two recessive homozygous pathogenic variants: in HERC2 and AP3B2. Known phenotypes related to HERC2 and AP3B2 include ID and early infantile epileptic encephalopathy, respectively. The patient had severe global developmental delay and profound ID and showed a happy demeanor, stereotypic laughter, and hand-flapping movements, but also irritability. Craniofacial dysmorphic features, including brachycephaly, strabismus, wide ala nasi, short philtrum, wide open mouth, and slight hypopigmentation were seen. Progressive microcephaly was noted. Magnetic resonance imaging of the brain showed delayed myelination and cerebral atrophy. Trio whole exome sequencing and CGH-SNP array analysis revealed paternal uniparental isodisomy of chromosome 15 and two coexisting recessive diseases resulting from homozygous HERC2 and AP3B2 pathogenic variants. The pathogenic variant in HERC2 was inherited from his heterozygous-carrier father, and the variant in AP3B2 was de novo. We suppose that these unusual features were the combination of the effect of three concomitant disorders.


Subject(s)
Adaptor Protein Complex 3/genetics , Adaptor Protein Complex beta Subunits/genetics , Angelman Syndrome/genetics , Intellectual Disability/genetics , Ubiquitin-Protein Ligases/genetics , Angelman Syndrome/diagnosis , Angelman Syndrome/pathology , Child, Preschool , Chromosomes, Human, Pair 15/genetics , Genetic Predisposition to Disease , Homozygote , Humans , Intellectual Disability/pathology , Male , Phenotype , Uniparental Disomy/genetics , Exome Sequencing
4.
Cerebellum ; 17(6): 709-721, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30046996

ABSTRACT

Synapses are precisely established, maintained, and modified throughout life by molecules called synaptic organizers, which include neurexins and neuroligins (Nlgn). Despite the importance of synaptic organizers in defining functions of neuronal circuits, the cellular and subcellular localization of many synaptic organizers has remained largely elusive because of the paucity of specific antibodies for immunohistochemical studies. In the present study, rather than raising specific antibodies, we generated knock-in mice in which a hemagglutinin (HA) epitope was inserted in the Nlgn1 gene. We have achieved high-throughput and precise gene editing by delivering the CRISPR/Cas9 system into zygotes. Using HA-Nlgn1 mice, we found that HA-Nlgn1 was enriched at synapses between parallel fibers and molecular layer interneurons (MLIs) and the glomeruli, in which mossy fiber terminals synapse onto granule cell dendrites. HA immunoreactivity was colocalized with postsynaptic density 95 at these synapses, indicating that endogenous Nlgn1 is localized at excitatory postsynaptic sites. In contrast, HA-Nlgn1 signals were very weak in dendrites and somata of Purkinje cells. Interestingly, HA-immunoreactivities were also observed in the pinceau, a specialized structure formed by MLI axons and astrocytes. HA-immunoreactivities in the pinceau were significantly reduced by knockdown of Nlgn1 in MLIs, indicating that in addition to postsynaptic sites, Nlgn1 is also localized at MLI axons. Our results indicate that epitope-tagging by electroporation-based gene editing with CRISPR/Cas9 is a viable and powerful method for mapping endogenous synaptic organizers with subcellular resolution, without the need for specific antibodies for each protein.


Subject(s)
Cell Adhesion Molecules, Neuronal/metabolism , Cerebellum/cytology , Cerebellum/metabolism , Animals , Astrocytes/cytology , Astrocytes/metabolism , CRISPR-Cas Systems , Cell Adhesion Molecules, Neuronal/genetics , Epitopes , Gene Knockdown Techniques , Genetic Engineering , HEK293 Cells , Hemagglutinins/genetics , Hemagglutinins/immunology , Hippocampus/cytology , Hippocampus/metabolism , Humans , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Inbred ICR , Mice, Transgenic , Neurons/cytology , Neurons/metabolism , Synapses/metabolism
5.
Biochem J ; 469(3): 445-54, 2015 Aug 01.
Article in English | MEDLINE | ID: mdl-26205497

ABSTRACT

Insulin-stimulated glucose uptake in skeletal muscle is mediated by the translocation of the glucose transporter GLUT4 from intracellular storage sites to the plasma membrane. The small GTPase Rac1 has been implicated in this insulin signalling, but the mechanism whereby Rac1 stimulates GLUT4 translocation remains obscure. In the present study, we examined the role of the small GTPase RalA downstream of Rac1 in skeletal muscle fibres isolated from genetically modified mice. A dominant-negative mutant of RalA, when ectopically overexpressed, significantly reduced GLUT4 translocation in response to insulin or either one of constitutively activated mutants of Rac1 and its upstream regulators, including the guanine-nucleotide-exchange factor FLJ00068, the protein kinase Akt2 and phosphoinositide 3-kinase. Constitutively activated Rac1 also failed to induce GLUT4 translocation in mouse skeletal muscle fibres in which the expression of RalA was abrogated by specific siRNA molecules. Furthermore, we applied a novel approach to detect the activated form of RalA in situ by immunofluorescence microscopy of mouse skeletal muscle fibres, demonstrating that constitutively activated mutants of Rac1 and its upstream regulators as well as insulin indeed cause the activation of RalA. Notably, this RalA activation was remarkably impaired in rac1-deficient skeletal muscle fibres. Taken together, these results provide evidence that RalA is indeed activated and involved in the regulation of GLUT4 translocation in response to insulin downstream of Rac1 in mouse skeletal muscle.


Subject(s)
Insulin/metabolism , Muscle, Skeletal/enzymology , Neuropeptides/metabolism , Signal Transduction , rac1 GTP-Binding Protein/metabolism , ral GTP-Binding Proteins/metabolism , Animals , Enzyme Activation , Glucose Transporter Type 4/genetics , Glucose Transporter Type 4/metabolism , Humans , Male , Mice , Mice, Inbred C57BL , Muscle Fibers, Skeletal/enzymology , Muscle Fibers, Skeletal/metabolism , Muscle, Skeletal/metabolism , Neuropeptides/genetics , rac1 GTP-Binding Protein/genetics , ral GTP-Binding Proteins/genetics
6.
J Neurosci ; 34(22): 7412-24, 2014 May 28.
Article in English | MEDLINE | ID: mdl-24872547

ABSTRACT

Of the two members of the δ subfamily of ionotropic glutamate receptors, GluD2 is exclusively expressed at parallel fiber-Purkinje cell (PF-PC) synapses in the cerebellum and regulates their structural and functional connectivity. However, little is known to date regarding cellular and synaptic expression of GluD1 and its role in synaptic circuit formation. In the present study, we investigated this issue by producing specific and sensitive histochemical probes for GluD1 and analyzing cerebellar synaptic circuits in GluD1-knock-out mice. GluD1 was widely expressed in the adult mouse brain, with high levels in higher brain regions, including the cerebral cortex, striatum, limbic regions (hippocampus, nucleus accumbens, lateral septum, bed nucleus stria terminalis, lateral habenula, and central nucleus of the amygdala), and cerebellar cortex. In the cerebellar cortex, GluD1 mRNA was expressed at the highest level in molecular layer interneurons and its immunoreactivity was concentrated at PF synapses on interneuron somata. In GluD1-knock-out mice, the density of PF synapses on interneuron somata was significantly reduced and the size and number of interneurons were significantly diminished. Therefore, GluD1 is common to GluD2 in expression at PF synapses, but distinct from GluD2 in neuronal expression in the cerebellar cortex; that is, GluD1 in interneurons and GluD2 in PCs. Furthermore, GluD1 regulates the connectivity of PF-interneuron synapses and promotes the differentiation and/or survival of molecular layer interneurons. These results suggest that GluD1 works in concert with GluD2 for the construction of cerebellar synaptic wiring through distinct neuronal and synaptic expressions and also their shared synapse-connecting function.


Subject(s)
Brain Chemistry/physiology , Cerebellum/physiology , Gene Expression Regulation/physiology , Interneurons/metabolism , Nerve Fibers, Myelinated/physiology , Receptors, Glutamate/biosynthesis , Synapses/physiology , Animals , Cell Differentiation/physiology , Cerebellum/ultrastructure , Glutamate Dehydrogenase , HEK293 Cells , Humans , Interneurons/ultrastructure , Mice , Mice, Inbred C57BL , Mice, Knockout , Nerve Fibers, Myelinated/ultrastructure , Nerve Net/physiology , Nerve Net/ultrastructure , Receptors, Glutamate/genetics , Receptors, Glutamate/physiology , Synapses/ultrastructure
7.
Vaccine ; 42(12): 3134-3143, 2024 Apr 30.
Article in English | MEDLINE | ID: mdl-38582691

ABSTRACT

OBJECTIVE: This study investigated the immunogenicity and safety of a pentavalent vaccine Gobik (DPT-IPV-Haemophilus influenzae type b [Hib]) in healthy Japanese infants aged ≥ 2 and < 43 months using a concomitant vaccination with ActHIB® (Hib) and Tetrabik (DPT-IPV) as a comparator. METHODS: This study was conducted as a phase 3, multicenter, active controlled, assessor-blinded, randomized, parallel-group study. Participants received a total of 4 subcutaneous doses (3 primary immunization doses and a booster dose) of either the experimental drug (DPT-IPV-Hib) or the active comparator (Hib + DPT-IPV). The primary endpoints were the anti-PRP antibody prevalence rate with ≥ 1 µg/mL, and the antibody prevalence rates against pertussis, diphtheria toxin, tetanus toxin, and attenuated poliovirus after the primary immunization. RESULTS: In 267 randomized participants (133 in the DPT-IPV-Hib group and 134 in the Hib + DPT-IPV group), the antibody prevalence rates after the primary immunization in both groups were 100.0 % and 88.7 % for anti-PRP antibody with ≥ 1 µg/mL, 99.2 % and 98.5 % against diphtheria toxin, and 100.0 % and 99.2 % against tetanus toxin, respectively. The antibody prevalence rates against pertussis and attenuated poliovirus were 100.0 % in both groups. The non-inferiority of the DPT-IPV-Hib group to the Hib + DPT-IPV group was verified for all measured antibodies. In both groups, all the GMTs of antibodies after the primary immunization were higher than those before the first dose, and those after the booster dose were higher than those after the primary immunization. No safety issues were identified. CONCLUSION: A single-agent Gobik, the first DPT-IPV-Hib pentavalent vaccine approved in Japan, was confirmed to simultaneously provide primary and booster immunizations against Hib infection, pertussis, diphtheria, tetanus, and poliomyelitis and to have a preventive effect and safety comparable to concomitant vaccination with Hib (ActHIB®) and DPT-IPV quadrivalent vaccine (Tetrabik).


Subject(s)
Diphtheria , Haemophilus Vaccines , Haemophilus influenzae type b , Poliomyelitis , Tetanus , Whooping Cough , Infant , Humans , Japan , Tetanus/prevention & control , Diphtheria/prevention & control , Whooping Cough/prevention & control , Tetanus Toxin , Diphtheria Toxin , Poliovirus Vaccine, Inactivated , Immunization Schedule , Antibodies, Bacterial , Diphtheria-Tetanus-Pertussis Vaccine , Vaccines, Combined , Poliomyelitis/prevention & control , Vaccines, Conjugate
8.
Genes (Basel) ; 15(3)2024 02 28.
Article in English | MEDLINE | ID: mdl-38540372

ABSTRACT

In newborn screening (NBS), it is important to consider the availability of multiplex assays or other tests that can be integrated into existing systems when attempting to implement NBS for new target diseases. Recent developments in innovative testing technology have made it possible to simultaneously screen for severe primary immunodeficiency (PID) and spinal muscular atrophy (SMA) using quantitative real-time polymerase chain reaction (qPCR) assays. We describe our experience of optional NBS for severe PID and SMA in Osaka, Japan. A multiplex TaqMan qPCR assay was used for the optional NBS program. The assay was able to quantify the levels of T-cell receptor excision circles and kappa-deleting recombination excision circles, which is useful for severe combined immunodeficiency and B-cell deficiency screening, and can simultaneously detect the homozygous deletion of SMN1 exon 7, which is useful for NBS for SMA. In total, 105,419 newborns were eligible for the optional NBS program between 1 August 2020 and 31 August 2023. A case each of X-linked agammaglobulinemia and SMA were diagnosed through the optional NBS and treated at early stages (before symptoms appeared). Our results show how multiplex PCR-based NBS can benefit large-scale NBS implementation projects for new target diseases.


Subject(s)
Muscular Atrophy, Spinal , Neonatal Screening , Infant, Newborn , Humans , Neonatal Screening/methods , Real-Time Polymerase Chain Reaction/methods , Homozygote , Japan , Sequence Deletion , Muscular Atrophy, Spinal/diagnosis , Muscular Atrophy, Spinal/genetics
9.
Mol Brain ; 16(1): 61, 2023 07 24.
Article in English | MEDLINE | ID: mdl-37488606

ABSTRACT

Changes in neural activity induced by learning and novel environments have been reported to lead to the formation of new synapses in the adult brain. However, the underlying molecular mechanism is not well understood. Here, we show that Purkinje cells (PCs), which have established adult-type monosynaptic innervation by climbing fibers (CFs) after elimination of weak CFs during development, can be reinnervated by multiple CFs by increased expression of the synaptic organizer C1ql1 in CFs or Bai3, a receptor for C1ql1, in PCs. In the adult cerebellum, CFs are known to have transverse branches that run in a mediolateral direction without forming synapses with PCs. Electrophysiological, Ca2+-imaging and immunohistochemical studies showed that overexpression of C1ql1 or Bai3 caused these CF transverse branches to elongate and synapse on the distal dendrites of mature PCs. Mature PCs were also reinnervated by multiple CFs when the glutamate receptor GluD2, which is essential for the maintenance of synapses between granule cells and PCs, was deleted. Interestingly, the effect of GluD2 knockout was not observed in Bai3 knockout PCs. In addition, C1ql1 levels were significantly upregulated in CFs of GluD2 knockout mice, suggesting that endogenous, not overexpressed, C1ql1-Bai3 signaling could regulate the reinnervation of mature PCs by CFs. Furthermore, the effects of C1ql1 and Bai3 overexpression required neuronal activity in the PC and CF, respectively. C1ql1 immunoreactivity at CF-PC synapses was reduced when the neuronal activity of CFs was suppressed. These results suggest that C1ql1-Bai3 signaling may mediate CF synaptogenesis in mature PCs, potentially in concert with neuronal activity.


Subject(s)
Neurons , Purkinje Cells , Animals , Mice , Dendrites , Cerebellum , Brain , Complement C1q
10.
Brain Dev ; 45(7): 363-371, 2023 Aug.
Article in English | MEDLINE | ID: mdl-36973114

ABSTRACT

OBJECTIVE: This study aimed to establish an optional newborn screening program for spinal muscular atrophy (SMA-NBS) in Osaka. METHODS: A multiplex TaqMan real-time quantitative polymerase chain reaction assay was used to screen for SMA. Dried blood spot samples obtained for the optional NBS program for severe combined immunodeficiency, which covers about 50% of the newborns in Osaka, were used. To obtain informed consent, participating obstetricians provided information about the optional NBS program to all parents by giving leaflets to prospective parents and uploading the information onto the internet. We prepared a workflow so that babies that were diagnosed with SMA through the NBS could be treated immediately. RESULTS: From 1 February 2021 to 30 September 2021, 22,951 newborns were screened for SMA. All of them tested negative for survival motor neuron (SMN)1 deletion, and there were no false-positives. Based on these results, an SMA-NBS program was established in Osaka and included in the optional NBS programs run in Osaka from 1 October 2021. A positive baby was found by screening, diagnosed with SMA (the baby possessed 3 copies of the SMN2 gene and was pre-symptomatic), and treated immediately. CONCLUSION: The workflow of the Osaka SMA-NBS program was confirmed to be useful for babies with SMA.


Subject(s)
Muscular Atrophy, Spinal , Neonatal Screening , Humans , Infant, Newborn , East Asian People , Muscular Atrophy, Spinal/diagnosis , Muscular Atrophy, Spinal/genetics , Neonatal Screening/methods , Pilot Projects , Prospective Studies , Survival of Motor Neuron 1 Protein/genetics , Japan
11.
Cerebellum ; 11(1): 78-84, 2012 Mar.
Article in English | MEDLINE | ID: mdl-20535596

ABSTRACT

Cerebellin was originally discovered as a Purkinje cell-specific peptide more than two decades ago. Later, its precursor protein precerebellin (Cbln1) was found to be produced in cerebellar granule cells. It has become increasingly clear that although the cerebellin peptide may have certain functions, Cbln1 is an actual signaling molecule that belongs to the C1q family. However, the precise function of Cbln1 has been unresolved. Cbln1 is released from granule cells, and disruption of the cbln1 gene in mice causes a severe reduction in the number of synapses between Purkinje cells and parallel fibers (PFs; axons of granule cells) and results in cerebellar ataxia. The glutamate receptor δ2 (GluD2) is highly expressed on Purkinje cells' dendritic spines which make synapses with PFs. Although GluD2 was identified as a member of the ionotropic glutamate receptors more than 15 years ago, it has been referred to as an orphan receptor because its endogenous ligands are unclear. Interestingly, GluD2-null mice phenocopy cbln1-null mice precisely. Cbln1 and GluD2 have therefore been thought to participate in a common signaling pathway that is required for the formation of PF synapses. We recently established a direct ligand-receptor relationship between Cbln1 and GluD2. The Cbln1-GluD2 complex is located at the cleft of PF-Purkinje cell synapses and bidirectionally regulates both presynaptic and postsynaptic differentiation.


Subject(s)
Cerebellar Cortex/growth & development , Ligand-Gated Ion Channels/physiology , Nerve Tissue Proteins/physiology , Protein Precursors/physiology , Receptors, Glutamate/physiology , Animals , Cell Differentiation/physiology , Cerebellar Cortex/cytology , Cerebellar Cortex/metabolism , Humans , Ligand-Gated Ion Channels/metabolism , Ligands , Mice , Nerve Tissue Proteins/metabolism , Presynaptic Terminals/metabolism , Presynaptic Terminals/physiology , Protein Precursors/metabolism , Receptors, Glutamate/metabolism , Synaptic Transmission/physiology
12.
J Neurosci ; 30(6): 2177-87, 2010 Feb 10.
Article in English | MEDLINE | ID: mdl-20147545

ABSTRACT

The lurcher (Lc) mice have served as a valuable model for neurodegeneration for decades. Although the responsible mutation was identified in genes encoding delta2 glutamate receptors (GluD2s), which are predominantly expressed in cerebellar Purkinje cells, how the mutant receptor (GluD2(Lc)) triggers cell death has remained elusive. Here, taking advantage of recent knowledge about the domain structure of GluD2, we reinvestigated Lc-mediated cell death, focusing on the "autophagic cell death" hypothesis. Although autophagy and cell death were induced by the expression of GluD2(Lc) in heterologous cells and cultured neurons, they were blocked by the introduction of mutations in the channel pore domain of GluD2(Lc) or by removal of extracellular Na(+). In addition, although GluD2(Lc) is reported to directly activate autophagy, mutant channels that are not associated with n-PIST (neuronal isoform of protein-interacting specifically with TC10)-Beclin1 still caused autophagy and cell death. Furthermore, cells expressing GluD2(Lc) showed decreased ATP levels and increased AMP-activated protein kinase (AMPK) activities in a manner dependent on extracellular Na(+). Thus, constitutive currents were likely necessary and sufficient to induce autophagy via AMPK activation, regardless of the n-PIST-Beclin1 pathway in vitro. Interestingly, the expression of dominant-negative AMPK suppressed GluD2(Lc)-induced autophagy but did not prevent cell death in heterologous cells. Similarly, the disruption of Atg5, a gene crucial for autophagy, did not prevent but rather aggravated Purkinje-cell death in Lc mice. Furthermore, calpains were specifically activated in Lc Purkinje cells. Together, these results suggest that Lc-mediated cell death was not caused by autophagy but necrosis with autophagic features both in vivo and in vitro.


Subject(s)
Autophagy , Calcium/metabolism , Nerve Degeneration/pathology , Neurons/pathology , Sodium/metabolism , AMP-Activated Protein Kinases/metabolism , Adaptor Proteins, Signal Transducing , Adenosine Triphosphate/metabolism , Animals , Apoptosis Regulatory Proteins/physiology , Autophagy-Related Protein 5 , Beclin-1 , Calpain/metabolism , Carrier Proteins/physiology , Cations , Cell Death , Cells, Cultured , Enzyme Activation , Golgi Matrix Proteins , Humans , Intracellular Signaling Peptides and Proteins , Mice , Mice, Neurologic Mutants , Microtubule-Associated Proteins/genetics , Nerve Degeneration/metabolism , Neurons/metabolism , Purkinje Cells/cytology , Purkinje Cells/metabolism , Receptors, Glutamate/biosynthesis , Receptors, Glutamate/genetics
13.
Eur J Neurosci ; 33(8): 1447-61, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21410790

ABSTRACT

Cbln1 (a.k.a. precerebellin) is a unique bidirectional synaptic organizer that plays an essential role in the formation and maintenance of excitatory synapses between granule cells and Purkinje cells in the mouse cerebellum. Cbln1 secreted from cerebellar granule cells directly induces presynaptic differentiation and indirectly serves as a postsynaptic organizer by binding to its receptor, the δ2 glutamate receptor. However, it remains unclear how Cbln1 binds to the presynaptic sites and interacts with other synaptic organizers. Furthermore, although Cbln1 and its family members Cbln2 and Cbln4 are expressed in brain regions other than the cerebellum, it is unknown whether they regulate synapse formation in these brain regions. In this study, we showed that Cbln1 and Cbln2, but not Cbln4, specifically bound to its presynaptic receptor -α and ß isoforms of neurexin carrying the splice site 4 insert [NRXs(S4+)] - and induced synaptogenesis in cerebellar, hippocampal and cortical neurons in vitro. Cbln1 competed with synaptogenesis mediated by neuroligin 1, which lacks the splice sites A and B, but not leucine-rich repeat transmembrane protein 2, possibly by sharing the presynaptic receptor NRXs(S4+). However, unlike neurexins/neuroligins or neurexins/leucine-rich repeat transmembrane proteins, the interaction between NRX1ß(S4+) and Cbln1 was insensitive to extracellular Ca(2+) concentrations. These findings revealed the unique and general roles of Cbln family proteins in mediating the formation and maintenance of synapses not only in the cerebellum but also in various other brain regions.


Subject(s)
Nerve Tissue Proteins/metabolism , Protein Isoforms/metabolism , Protein Precursors/metabolism , Signal Transduction/physiology , Synapses/physiology , Animals , Cell Differentiation , Cells, Cultured , Cerebellum/cytology , Glycoproteins/genetics , Glycoproteins/metabolism , HEK293 Cells , Hippocampus/cytology , Humans , Mice , Nerve Tissue Proteins/genetics , Neuropeptides/genetics , Neuropeptides/metabolism , Protein Binding , Protein Isoforms/genetics , Protein Precursors/genetics , Receptors, Glutamate/genetics , Receptors, Glutamate/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Synapses/ultrastructure
14.
Am J Med Genet A ; 155A(8): 1949-58, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21744491

ABSTRACT

Loss-of-function mutations in CHST14, dermatan 4-O-sulfotransferase 1 (D4ST1) deficiency, have recently been found to cause adducted thumb-clubfoot syndrome (ATCS; OMIM#601776) and a new type of Ehlers-Danlos syndrome (EDS) coined as EDS Kosho Type (EDSKT) [Miyake et al., 2010], as well as a subset of kyphoscoliosis type EDS without lysyl hydroxylase deficiency (EDS VIB) coined as musculocontractural EDS (MCEDS) [Malfait et al., 2010]. Lack of detailed clinical information from later childhood to adulthood in ATCS and lack of detailed clinical information from birth to early childhood in EDSKT and MCEDS have made it difficult to determine whether these disorders would be distinct clinical entities or a single clinical entity with variable expressions and with different presentations depending on the patients' ages at diagnosis. We present detailed clinical findings and courses of two additional unrelated patients, aged 2 years and 6 years, with EDSKT with a comprehensive review of 20 reported patients with D4ST1 deficiency, which supports the notion that these disorders constitute a clinically recognizable form of EDS. The disorder, preferably termed D4ST1-deficient EDS, is characterized by progressive multisystem fragility-related manifestations (joint dislocations and deformities, skin hyperextensibility, bruisability, and fragility; recurrent large subcutaneous hematomas, and other cardiac valvular, respiratory, gastrointestinal, and ophthalmological complications) resulting from impaired assembly of collagen fibrils, as well as various malformations (distinct craniofacial features, multiple congenital contractures, and congenital defects in cardiovascular, gastrointestinal, renal, ocular, and central nervous systems) resulting from inborn errors of development.


Subject(s)
Abnormalities, Multiple/genetics , Ehlers-Danlos Syndrome/enzymology , Sulfotransferases/deficiency , Child , Child, Preschool , Clubfoot/surgery , Craniofacial Abnormalities/genetics , Cryptorchidism/genetics , DNA Mutational Analysis , Developmental Disabilities/genetics , Ehlers-Danlos Syndrome/genetics , Genetic Association Studies , Humans , Male , Phenotype , Sulfotransferases/genetics
15.
J Neurosci ; 29(18): 5738-48, 2009 May 06.
Article in English | MEDLINE | ID: mdl-19420242

ABSTRACT

The delta2 glutamate receptor (GluRdelta2; GluD2), which is predominantly expressed on postsynaptic sites at parallel fiber (PF)-Purkinje cell synapses in the cerebellum, plays two crucial roles in the cerebellum: the formation of PF synapses and the regulation of long-term depression (LTD), a form of synaptic plasticity underlying motor learning. Although the induction of LTD and motor learning absolutely require signaling via the cytoplasmic C-terminal domain of GluD2, the mechanisms by which GluD2 regulates PF synaptogenesis have remained unclear. Here, we examined the role of the extracellular N-terminal domain (NTD) of GluD2 on PF synaptogenesis by injecting Sindbis virus carrying wild-type (GluD2(wt)) or mutant GluD2 into the subarachnoid supracerebellar space of GluD2-null mice. Remarkably, the expression of GluD2(wt), but not of a mutant GluD2 lacking the NTD (GluD2(DeltaNTD)), rapidly induced PF synapse formation and rescued gross motor dyscoordination in adult GluD2-null mice just 1 d after injection. In addition, although the kainate receptor GluR6 (GluK2) did not induce PF synaptogenesis, a chimeric GluK2 that contained the NTD of GluD2 (GluD2(NTD)-GluK2) did. Similarly, GluD2(wt) and GluD2(NTD)-GluK2, but not GluD2(DeltaNTD), induced synaptogenesis in heterologous cells in vitro. In contrast, LTD was restored in GluD2-null Purkinje cells expressing a mutant GluD2 lacking the NTD. These results indicate that the NTD of GluD2 is necessary and sufficient for the function of GluD2 in the regulation of PF-Purkinje cell synaptogenesis. Furthermore, our results suggest that GluD2 differently regulates PF synaptogenesis and cerebellar LTD through the extracellular NTD and the cytoplasmic C-terminal end, respectively.


Subject(s)
Cerebellum/cytology , Presynaptic Terminals/physiology , Protein Structure, Tertiary/physiology , Receptors, Glutamate/chemistry , Synapses/metabolism , Amino Acid Sequence/genetics , Analysis of Variance , Animals , Animals, Newborn , Cell Line, Transformed , Cerebellum/physiology , Disease Models, Animal , Electric Stimulation/methods , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/genetics , Gene Expression/genetics , Gene Transfer Techniques , Green Fluorescent Proteins/genetics , Humans , Long-Term Synaptic Depression/genetics , Mice , Mice, Knockout , Microscopy, Electron, Transmission , Models, Molecular , Motor Activity/genetics , Motor Skills Disorders/genetics , Motor Skills Disorders/therapy , Mutation/genetics , Nerve Fibers/physiology , Neuronal Plasticity , Patch-Clamp Techniques , Presynaptic Terminals/ultrastructure , Protein Structure, Tertiary/genetics , Protein Transport/genetics , Purkinje Cells/cytology , Purkinje Cells/drug effects , Purkinje Cells/ultrastructure , Receptors, Glutamate/deficiency , Receptors, Glutamate/genetics , Recruitment, Neurophysiological , Sindbis Virus/physiology , Synapses/drug effects , Synapses/genetics , Synapses/ultrastructure , Transfection/methods , Vesicular Glutamate Transport Protein 1/metabolism
16.
Am J Med Genet A ; 152A(8): 2103-9, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20635407

ABSTRACT

A patient with atypical phenotypes of Prader-Willi syndrome (PWS) was subjected to investigate genomic copy numbers by microarray-based comparative genomic hybridization analysis. Severe developmental delay, relative macrocephaly, protruding forehead, cardiac anomalies, and hydronephrosis were atypical for PWS. Concurrent deletions of 15q11-13 and 5q35 regions were revealed and identified as paternally derived. The sizes and locations of the two deletions were typical for both deletions. Although each deletion independently contributed to the clinical features, developmental disturbance was very severe, suggesting combined effects. This is the first report of co-occurrence of PWS and STS. The co-occurrence of two syndromes is likely incidental.


Subject(s)
Chromosomes, Human, Pair 15/genetics , Developmental Disabilities/genetics , Prader-Willi Syndrome/genetics , Adolescent , Comparative Genomic Hybridization , Developmental Disabilities/complications , Humans , In Situ Hybridization, Fluorescence , Male , Oligonucleotide Array Sequence Analysis , Prader-Willi Syndrome/complications , Syndrome
17.
Science ; 369(6507)2020 08 28.
Article in English | MEDLINE | ID: mdl-32855309

ABSTRACT

Neuronal synapses undergo structural and functional changes throughout life, which are essential for nervous system physiology. However, these changes may also perturb the excitatory-inhibitory neurotransmission balance and trigger neuropsychiatric and neurological disorders. Molecular tools to restore this balance are highly desirable. Here, we designed and characterized CPTX, a synthetic synaptic organizer combining structural elements from cerebellin-1 and neuronal pentraxin-1. CPTX can interact with presynaptic neurexins and postsynaptic AMPA-type ionotropic glutamate receptors and induced the formation of excitatory synapses both in vitro and in vivo. CPTX restored synaptic functions, motor coordination, spatial and contextual memories, and locomotion in mouse models for cerebellar ataxia, Alzheimer's disease, and spinal cord injury, respectively. Thus, CPTX represents a prototype for structure-guided biologics that can efficiently repair or remodel neuronal circuits.


Subject(s)
C-Reactive Protein/pharmacology , Nerve Tissue Proteins/pharmacology , Neural Pathways/drug effects , Protein Precursors/pharmacology , Receptors, AMPA/metabolism , Recombinant Proteins/pharmacology , Synapses/drug effects , Alzheimer Disease/therapy , Animals , C-Reactive Protein/chemistry , C-Reactive Protein/therapeutic use , Cerebellar Ataxia/therapy , Disease Models, Animal , HEK293 Cells , Hippocampus , Humans , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/therapeutic use , Protein Domains , Protein Precursors/chemistry , Protein Precursors/therapeutic use , Receptors, Glutamate/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/therapeutic use , Spine/drug effects , Spine/physiology
18.
J Neurosci ; 28(6): 1460-8, 2008 Feb 06.
Article in English | MEDLINE | ID: mdl-18256267

ABSTRACT

The delta2 glutamate receptor (GluRdelta2) is predominantly expressed in Purkinje cells and plays crucial roles in cerebellar functions: GluRdelta2-/- mice display ataxia and impaired motor learning. In addition, long-term depression (LTD) at parallel fiber (PF)-Purkinje cell synapses is abrogated, and synapse formation with PFs and climbing fibers (CFs) is severely disturbed in GluRdelta2-/- Purkinje cells. Recently, we demonstrated that abrogated LTD was restored in GluRdelta2-/- Purkinje cells by the virus-mediated expression of the wild-type GluRdelta2 transgene (Tg(wt)) but not by that of mutant GluRdelta2 lacking the C-terminal seven residues to which several PDZ proteins bind (Tg(DeltaCT7)). These results indicated that the C terminus of GluRdelta2 conveys the signal(s) necessary for LTD. In contrast, other phenotypes of GluRdelta2-/- cerebellum, especially morphological abnormalities at PF and CF synapses, could not be rescued by virus-mediated transient expression. Thus, whether these phenotypes are mediated by the same signaling pathway remains unclear. To address these issues and to further delineate the function of GluRdelta2 in vivo, we generated transgenic mice that expressed Tg(DeltaCT7) on a GluRdelta2-/- background. Interestingly, although Tg(DeltaCT7) restored abnormal PF and CF synapse formation almost completely, it could not rescue abrogated LTD in GluRdelta2-/- Purkinje cells. Furthermore, although the gross motor discoordination of GluRdelta2-/- mice was restored, the cerebellar motor learning underlying delayed eyeblink conditioning remained impaired. These results indicate that LTD induction and motor learning are regulated by signaling via the C-terminal end of GluRdelta2, whereas other functions may be differentially regulated by other regions of GluRdelta2.


Subject(s)
Learning/physiology , Motor Skills/physiology , Neuronal Plasticity/physiology , PDZ Domains/physiology , Peptide Fragments/physiology , Receptors, Glutamate/physiology , Synapses/metabolism , Animals , Cerebellum/physiology , Excitatory Postsynaptic Potentials/genetics , Long-Term Synaptic Depression/genetics , Male , Mice , Mice, Transgenic , PDZ Domains/genetics , Peptide Fragments/biosynthesis , Peptide Fragments/genetics , Protein Binding/genetics , Receptors, Glutamate/biosynthesis , Receptors, Glutamate/genetics , Synapses/genetics
19.
J Neurosci ; 28(23): 5920-30, 2008 Jun 04.
Article in English | MEDLINE | ID: mdl-18524896

ABSTRACT

Although many synapse-organizing molecules have been identified in vitro, their functions in mature neurons in vivo have been mostly unexplored. Cbln1, which belongs to the C1q/tumor necrosis factor superfamily, is the most recently identified protein involved in synapse formation in the mammalian CNS. In the cerebellum, Cbln1 is predominantly produced and secreted from granule cells; cbln1-null mice show ataxia and a severe reduction in the number of synapses between Purkinje cells and parallel fibers (PFs), the axon bundle of granule cells. Here, we show that application of recombinant Cbln1 specifically and reversibly induced PF synapse formation in dissociated cbln1-null Purkinje cells in culture. Cbln1 also rapidly induced electrophysiologically functional and ultrastructurally normal PF synapses in acutely prepared cbln1-null cerebellar slices. Furthermore, a single injection of recombinant Cbln1 rescued severe ataxia in adult cbln1-null mice in vivo by completely, but transiently, restoring PF synapses. Therefore, Cbln1 is a unique synapse organizer that is required not only for the normal development of PF-Purkinje cell synapses but also for their maintenance in the mature cerebellum both in vitro and in vivo. Furthermore, our results indicate that Cbln1 can also rapidly organize new synapses in adult cerebellum, implying its therapeutic potential for cerebellar ataxic disorders.


Subject(s)
Excitatory Postsynaptic Potentials/physiology , Nerve Tissue Proteins/physiology , Protein Precursors/physiology , Purkinje Cells/physiology , Synapses/physiology , Age Factors , Animals , Cell Line , Cells, Cultured , Cerebellum/growth & development , Cerebellum/ultrastructure , Excitatory Postsynaptic Potentials/genetics , Mice , Mice, Knockout , Nerve Tissue Proteins/deficiency , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/pharmacology , Protein Precursors/deficiency , Protein Precursors/genetics , Protein Precursors/pharmacology , Purkinje Cells/metabolism , Purkinje Cells/ultrastructure , Synapses/genetics , Synapses/ultrastructure
20.
Eur J Neurosci ; 29(4): 707-17, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19200061

ABSTRACT

Cbln1, which belongs to the C1q/tumor necrosis factor superfamily, is a unique molecule that is not only required for maintaining normal parallel fiber (PF)-Purkinje cell synapses, but is also capable of inducing new PF synapses in adult cerebellum. Although Cbln1 is reportedly released from granule cells, where and how Cbln1 binds in the cerebellum has remained largely unclear, partly because Cbln1 undergoes proteolysis to yield various fragments that are differentially detected by different antibodies. To circumvent this problem, we characterized the Cbln1-binding site using recombinant Cbln1. An immunohistochemical analysis revealed that recombinant Cbln1 preferentially bound to PF-Purkinje cell synapses in primary cultures and acute slice preparations in a saturable and replaceable manner. Specific binding was observed for intact Cbln1 that had formed a hexamer, but not for the N-terminal or C-terminal fragments of Cbln1 fused to other proteins. Similarly, mutant Cbln1 that had formed a trimer did not bind to the Purkinje cells. Immunoreactivity for the recombinant Cbln1 was observed in weaver cerebellum (which lacks granule cells) but was absent in pcd cerebellum (which lacks Purkinje cells), suggesting that the binding site was located on the postsynaptic sites of PF-Purkinje cell synapses. Finally, a subcellular fractionation analysis revealed that recombinant Cbln1 bound to the synaptosomal and postsynaptic density fractions. These results indicate that Cbln1, released from granule cells as hexamers, specifically binds to a putative receptor located at the postsynaptic sites of PF-Purkinje cell synapses, where it induces synaptogenesis.


Subject(s)
Cerebellum/metabolism , Nerve Tissue Proteins/metabolism , Protein Precursors/metabolism , Purkinje Cells/metabolism , Synapses/metabolism , Animals , Cell Line , Cells, Cultured , Dendrites/metabolism , Humans , Immunohistochemistry , Mice , Mice, Inbred ICR , Mice, Knockout , Mice, Neurologic Mutants , Microscopy, Confocal , Mutation , Nerve Tissue Proteins/genetics , Protein Precursors/genetics , Synaptosomes/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL