Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
1.
Int J Mol Sci ; 24(23)2023 Dec 04.
Article in English | MEDLINE | ID: mdl-38069416

ABSTRACT

Mammalian auditory hair cells transduce sound-evoked traveling waves in the cochlea into nerve stimuli, which are essential for hearing function. Pillar cells located between the inner and outer hair cells are involved in the formation of the tunnel of Corti, which incorporates outer-hair-cell-driven fluid oscillation and basilar membrane movement, leading to the fine-tuned frequency-specific perception of sounds by the inner hair cells. However, the detailed molecular mechanism underlying the development and maintenance of pillar cells remains to be elucidated. In this study, we examined the expression and function of brain-specific angiogenesis inhibitor 3 (Bai3), an adhesion G-protein-coupled receptor, in the cochlea. We found that Bai3 was expressed in hair cells in neonatal mice and pillar cells in adult mice, and, interestingly, Bai3 knockout mice revealed the abnormal formation of pillar cells, with the elevation of the hearing threshold in a frequency-dependent manner. Furthermore, old Bai3 knockout mice showed the degeneration of hair cells and spiral ganglion neurons in the basal turn. The results suggest that Bai3 plays a crucial role in the development and/or maintenance of pillar cells, which, in turn, are necessary for normal hearing function. Our results may contribute to understanding the mechanisms of hearing loss in human patients.


Subject(s)
Cochlea , Hearing , Membrane Proteins , Nerve Tissue Proteins , Animals , Mice , Brain , Cochlea/metabolism , Hair Cells, Auditory, Outer , Mice, Knockout , Nerve Tissue Proteins/genetics , Membrane Proteins/genetics
2.
EMBO J ; 36(9): 1227-1242, 2017 05 02.
Article in English | MEDLINE | ID: mdl-28283581

ABSTRACT

The axon initial segment (AIS) is a specialized domain essential for neuronal function, the formation of which begins with localization of an ankyrin-G (AnkG) scaffold. However, the mechanism directing and maintaining AnkG localization is largely unknown. In this study, we demonstrate that in vivo knockdown of microtubule cross-linking factor 1 (MTCL1) in cerebellar Purkinje cells causes loss of axonal polarity coupled with AnkG mislocalization. MTCL1 lacking MT-stabilizing activity failed to restore these defects, and stable MT bundles spanning the AIS were disorganized in knockdown cells. Interestingly, during early postnatal development, colocalization of MTCL1 with these stable MT bundles was observed prominently in the axon hillock and proximal axon. These results indicate that MTCL1-mediated formation of stable MT bundles is crucial for maintenance of AnkG localization. We also demonstrate that Mtcl1 gene disruption results in abnormal motor coordination with Purkinje cell degeneration, and provide evidence suggesting possible involvement of MTCL1 dysfunction in the pathogenesis of spinocerebellar ataxia.


Subject(s)
Axon Initial Segment/physiology , Microtubule-Associated Proteins/metabolism , Purkinje Cells/cytology , Purkinje Cells/physiology , Animals , Gene Knockdown Techniques , Gene Knockout Techniques , Mice , Mice, Knockout , Motor Disorders
3.
J Neurosci ; 35(36): 12518-34, 2015 Sep 09.
Article in English | MEDLINE | ID: mdl-26354918

ABSTRACT

The establishment of cell-type-specific dendritic arbors is fundamental for proper neural circuit formation. Here, using temporal- and cell-specific knock-down, knock-out, and overexpression approaches, we show that multiple aspects of the dendritic organization of cerebellar Purkinje cells (PCs) are controlled by a single transcriptional factor, retinoic acid-related orphan receptor-alpha (RORα), a gene defective in staggerer mutant mice. As reported earlier, RORα was required for regression of primitive dendrites before postnatal day 4 (P4). RORα was also necessary for PCs to form a single Purkinje layer from P0 to P4. The knock-down of RORα from P4 impaired the elimination of perisomatic dendrites and maturation of single stem dendrites in PCs at P8. Filopodia and spines were also absent in these PCs. The knock-down of RORα from P8 impaired the formation and maintenance of terminal dendritic branches of PCs at P14. Finally, even after dendrite formation was completed at P21, RORα was required for PCs to maintain dendritic complexity and functional synapses, but their mature innervation pattern by single climbing fibers was unaffected. Interestingly, overexpression of RORα in PCs at various developmental stages did not facilitate dendrite development, but had specific detrimental effects on PCs. Because RORα deficiency during development is closely related to the severity of spinocerebellar ataxia type 1, delineating the specific roles of RORα in PCs in vivo at different time windows during development and throughout adulthood would facilitate our understanding of the pathogenesis of cerebellar disorders. Significance statement: The genetic programs by which each neuron subtype develops and maintains dendritic arbors have remained largely unclear. This is partly because dendrite development is modulated dynamically by neuronal activities and interactions with local environmental cues in vivo. In addition, dendrites are formed and maintained by the balance between their growth and regression; the effects caused by the disruption of transcription factors during the early developmental stages could be masked by dendritic growth or regression in the later stages. Here, using temporal- and cell-specific knock-down, knock-out, and overexpression approaches in vivo, we show that multiple aspects of the dendritic organization of cerebellar Purkinje cells are controlled by a single transcriptional factor, retinoic acid-related orphan receptor alpha.


Subject(s)
Dendrites/metabolism , Neurogenesis , Nuclear Receptor Subfamily 1, Group F, Member 1/metabolism , Purkinje Cells/metabolism , Animals , Dendrites/physiology , Mice , Mice, Inbred ICR , Mice, Neurologic Mutants , Nuclear Receptor Subfamily 1, Group F, Member 1/genetics , Purkinje Cells/cytology
4.
Eur J Neurosci ; 39(8): 1268-80, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24467251

ABSTRACT

The formation of excitatory and inhibitory synapses must be tightly coordinated to establish functional neuronal circuitry during development. In the cerebellum, the formation of excitatory synapses between parallel fibers and Purkinje cells is strongly induced by Cbln1, which is released from parallel fibers and binds to the postsynaptic δ2 glutamate receptor (GluD2). However, Cbln1's role, if any, in inhibitory synapse formation has been unknown. Here, we show that Cbln1 downregulates the formation and function of inhibitory synapses between Purkinje cells and interneurons. Immunohistochemical analyses with an anti-vesicular GABA transporter antibody revealed an increased density of interneuron-Purkinje cell synapses in the cbln1-null cerebellum. Whole-cell patch-clamp recordings from Purkinje cells showed that both the amplitude and frequency of miniature inhibitory postsynaptic currents were increased in cbln1-null cerebellar slices. A 3-h incubation with recombinant Cbln1 reversed the increased amplitude of inhibitory currents in Purkinje cells in acutely prepared cbln1-null slices. Furthermore, an 8-day incubation with recombinant Cbln1 reversed the increased interneuron-Purkinje cell synapse density in cultured cbln1-null slices. In contrast, recombinant Cbln1 did not affect cerebellar slices from mice lacking both Cbln1 and GluD2. Finally, we found that tyrosine phosphorylation was upregulated in the cbln1-null cerebellum, and acute inhibition of Src-family kinases suppressed the increased inhibitory postsynaptic currents in cbln1-null Purkinje cells. These findings indicate that Cbln1-GluD2 signaling inhibits the number and function of inhibitory synapses, and shifts the excitatory-inhibitory balance towards excitation in Purkinje cells. Cbln1's effect on inhibitory synaptic transmission is probably mediated by a tyrosine kinase pathway.


Subject(s)
Inhibitory Postsynaptic Potentials , Nerve Tissue Proteins/metabolism , Protein Precursors/metabolism , Purkinje Cells/metabolism , Synapses/metabolism , Animals , Down-Regulation , Interneurons/metabolism , Interneurons/physiology , Mice , Miniature Postsynaptic Potentials , Nerve Tissue Proteins/genetics , Neurogenesis , Protein Precursors/genetics , Purkinje Cells/cytology , Purkinje Cells/physiology , Receptors, Glutamate/genetics , Receptors, Glutamate/metabolism , Synapses/physiology , src-Family Kinases/metabolism
5.
Chem ; 9(2): 523-540, 2023 Feb.
Article in English | MEDLINE | ID: mdl-38094901

ABSTRACT

Various small molecules have been used as functional probes for tissue imaging in medical diagnosis and pharmaceutical drugs for disease treatment. The spatial distribution, target selectivity, and diffusion/excretion kinetics of small molecules in structurally complicated specimens are critical for function. However, robust methods for precisely evaluating these parameters in the brain have been limited. Herein, we report a new method termed "fixation-driven chemical cross-linking of exogenous ligands (FixEL)," which traps and images exogenously administered molecules of interest (MOIs) in complex tissues. This method relies on protein-MOI interactions and chemical cross-linking of amine-tethered MOI with paraformaldehyde used for perfusion fixation. FixEL is used to obtain images of the distribution of the small molecules, which addresses selective/nonselective binding to proteins, time-dependent localization changes, and diffusion/retention kinetics of MOIs such as the scaffold of PET tracer derivatives or drug-like small molecules.

6.
Eur J Neurosci ; 36(7): 2867-76, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22775058

ABSTRACT

Cerebellar Purkinje cells, which convey the only output from the cerebellar cortex, play an essential role in cerebellar functions, such as motor coordination and motor learning. To understand how Purkinje cells develop and function in the mature cerebellum, an efficient method for molecularly perturbing them is needed. Here we demonstrate that Purkinje cell progenitors at embryonic day (E)11.5 could be efficiently and preferentially transfected by spatially directed in utero electroporation (IUE) with an optimized arrangement of electrodes. Electrophysiological analyses indicated that the electroporated Purkinje cells maintained normal membrane properties, synaptic responses and synaptic plasticity at postnatal days 25-28. By combining the L7 promoter and inducible Cre/loxP system with IUE, transgenes were expressed even more specifically in Purkinje cells and in a temporally controlled manner. We also show that three different fluorescent proteins could be simultaneously expressed, and that Bassoon, a large synaptic protein, could be expressed in the electroporated Purkinje cells. Moreover, phenotypes of staggerer mutant mice, which have a deletion in the gene encoding retinoid-related orphan receptor α (RORα1), were recapitulated by electroporating a dominant-negative form of RORα1 into Purkinje cells at E11.5. Together, these results indicate that this new IUE protocol, which allows the selective, effective and temporally regulated expression of multiple foreign genes transfected into Purkinje cell progenitors in vivo, without changing the cells' physiological characteristics, is a powerful tool for elucidating the molecular mechanisms underlying early Purkinje cell developmental events, such as dendritogenesis and migration, and synaptic plasticity in mature Purkinje cells.


Subject(s)
Electroporation/methods , Gene Expression Regulation, Developmental , Purkinje Cells/metabolism , Animals , Embryonic Stem Cells/metabolism , Female , Genetic Vectors/genetics , Membrane Potentials/genetics , Mice , Mice, Neurologic Mutants , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neural Stem Cells/metabolism , Nuclear Receptor Subfamily 1, Group F, Member 1/genetics , Nuclear Receptor Subfamily 1, Group F, Member 1/metabolism , Pregnancy , Promoter Regions, Genetic/genetics , Purkinje Cells/physiology , Synaptic Transmission/genetics , Transfection , Transgenes/genetics
7.
Plant Physiol ; 155(3): 1214-25, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21205615

ABSTRACT

The secondary growth of a woody stem requires the formation of a vascular cambium at an appropriate position and proper patterning of the vascular tissues derived from the cambium. Class III homeodomain-leucine zipper (HD ZIP) transcription factors have been implicated in polarity determination and patterning in lateral organs and primary vascular tissues and in the initiation and function of shoot apical meristems. We report here the functional characterization of a Populus class III HD ZIP gene, popREVOLUTA (PRE), that demonstrates another role for class III HD ZIPs in regulating the development of cambia and secondary vascular tissues. PRE is orthologous to Arabidopsis (Arabidopsis thaliana) REVOLUTA and is expressed in both the shoot apical meristem and in the cambial zone and secondary vascular tissues. Transgenic Populus expressing a microRNA-resistant form of PRE presents unstable phenotypic abnormalities affecting both primary and secondary growth. Surprisingly, phenotypic changes include abnormal formation of cambia within cortical parenchyma that can produce secondary vascular tissues in reverse polarity. Genes misexpressed in PRE mutants include transcription factors and auxin-related genes previously implicated in class III HD ZIP functions during primary growth. Together, these results suggest that PRE plays a fundamental role in the initiation of the cambium and in regulating the patterning of secondary vascular tissues.


Subject(s)
Body Patterning , Cambium/embryology , Homeodomain Proteins/metabolism , Leucine Zippers/genetics , Plant Proteins/metabolism , Plant Stems/embryology , Populus/embryology , Cambium/cytology , Cambium/genetics , Cambium/growth & development , Cell Differentiation/genetics , Gene Expression Regulation, Developmental , Gene Expression Regulation, Plant , Genes, Plant/genetics , Homeodomain Proteins/genetics , MicroRNAs/metabolism , Mutation/genetics , Organ Specificity , Phenotype , Phylogeny , Plant Growth Regulators/genetics , Plant Proteins/genetics , Plant Stems/cytology , Plant Stems/genetics , Plant Vascular Bundle/genetics , Plant Vascular Bundle/growth & development , Plants, Genetically Modified , Populus/cytology , Populus/genetics , Sequence Homology, Amino Acid , Wood/cytology , Wood/genetics , Wood/growth & development
8.
Neurosci Res ; 180: 13-22, 2022 Jul.
Article in English | MEDLINE | ID: mdl-35247520

ABSTRACT

Calcineurin (CN) is a Ca2+/calmodulin-dependent serine/threonine protein phosphatase consisting of catalytic CNA and regulatory CNB subunits, and links activity-dependent Ca2+ signals to various neural functions. Here we studied CN expression in the mouse brain by producing subunit-specific probes and antibodies. Of five CN subunits. CNAα, CNAß, and CNB1 mRNAs were predominantly expressed over the brain from early embryonic to adult stage, and all were high in the telencephalon and cerebellum. Protein localization was examined in the cerebellum by immunofluorescence with cellular and terminal markers and by preembedding silver-enhanced immunogold microscopy. CNB1 and CNAß were co-distributed in subcellular and synaptic elements of various cerebellar neurons and glia, whereas CNAα was exclusive in granule cell elements, including parallel fiber terminals. The present study thus discloses that CNB1 subunit well coexists with one or two CNA subunits in various cerebellar compartments. Moreover, high CN contents are provided to parallel fiber-Purkinje cell synapses, i.e., CNAα, CNAß, and CNB1 in their presynaptic side and CNAß and CNB1 in their postsynaptic side. These findings will be the anatomical basis, at least partly, for the known regulatory roles of postsynaptic CNs in long-term depression and presynaptic CNs in transmitter release function.


Subject(s)
Calcineurin , Purkinje Cells , Animals , Calcineurin/metabolism , Cerebellum/physiology , Mice , Neuroglia/metabolism , Neurons/metabolism , Purkinje Cells/physiology , Synapses/physiology
9.
Neuron ; 110(19): 3168-3185.e8, 2022 10 05.
Article in English | MEDLINE | ID: mdl-36007521

ABSTRACT

Excitatory synapses are formed and matured by the cooperative actions of synaptic organizers, such as neurexins (Nrxns), neuroligins (Nlgns), LRRTMs, and Cbln1. Recent super-resolution nanoscopy developments have revealed that many synaptic organizers, as well as glutamate receptors and glutamate release machinery, exist as nanoclusters within synapses. However, it is unclear how such nanodomains interact with each other to organize excitatory synapses in vivo. By applying X10 expansion microscopy to epitope tag knockin mice, we found that Cbln1, Nlgn1, and LRRTM1, which share Nrxn as a common presynaptic receptor, form overlapping or separate nanodomains depending on Nrxn with or without a sequence encoded by splice site 4. The size and position of glutamate receptor nanodomains of GluD1, NMDA, and AMPA receptors were regulated by Cbln1, Nlgn1, and LRRTM1 nanodomains, respectively. These findings indicate that Nrxns anterogradely regulate the postsynaptic nanoscopic architecture of glutamate receptors through competition and coordination of Nrxn ligands.


Subject(s)
Nerve Tissue Proteins , Receptors, AMPA , Animals , Cell Adhesion Molecules, Neuronal/genetics , Cell Adhesion Molecules, Neuronal/metabolism , Epitopes , Glutamic Acid , Ligands , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , N-Methylaspartate , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Receptors, Glutamate/genetics , Receptors, Glutamate/metabolism , Receptors, Presynaptic , Synapses/physiology
10.
Nat Commun ; 13(1): 3167, 2022 06 16.
Article in English | MEDLINE | ID: mdl-35710788

ABSTRACT

Direct activation of cell-surface receptors is highly desirable for elucidating their physiological roles. A potential approach for cell-type-specific activation of a receptor subtype is chemogenetics, in which both point mutagenesis of the receptors and designed ligands are used. However, ligand-binding properties are affected in most cases. Here, we developed a chemogenetic method for direct activation of metabotropic glutamate receptor 1 (mGlu1), which plays essential roles in cerebellar functions in the brain. Our screening identified a mGlu1 mutant, mGlu1(N264H), that was activated directly by palladium complexes. A palladium complex showing low cytotoxicity successfully activated mGlu1 in mGlu1(N264H) knock-in mice, revealing that activation of endogenous mGlu1 is sufficient to evoke the critical cellular mechanism of synaptic plasticity, a basis of motor learning in the cerebellum. Moreover, cell-type-specific activation of mGlu1 was demonstrated successfully using adeno-associated viruses in mice, which shows the potential utility of this chemogenetics for clarifying the physiological roles of mGlu1 in a cell-type-specific manner.


Subject(s)
Cerebellum , Palladium , Animals , Brain , Mice , Neuronal Plasticity
11.
Plant J ; 64(3): 536-50, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20807213

ABSTRACT

In yeast, eIF5A, in combination with eEF2, functions at the translation step, during the protein elongation cycle. This result is of significance with respect to functioning of the enucleate sieve tube system, as eIF5A was recently detected in Cucurbita maxima (pumpkin) phloem sap. In the present study, we further characterized four CmeIF5A isoforms, encoding three proteins, all of which were present in the phloem sap. Although hypusination of CmeIF5A was not necessary for entry into the sieve elements, this unique post-translational modification was necessary for RNA binding. The two enzymes required for hypusination were detected in pumpkin phloem sap, where presumably this modification takes place. A combination of gel-filtration chromatography and protein overlay assays demonstrated that, as in yeast, CmeIF5A interacts with phloem proteins, like eEF2, known to be involved in protein synthesis. These findings are discussed in terms of a potential role for eIF5A in regulating protein synthesis within the enucleate sieve tube system of the angiosperms.


Subject(s)
Cucurbita/genetics , Peptide Initiation Factors/metabolism , Phloem/metabolism , Plant Proteins/metabolism , Protein Biosynthesis , RNA-Binding Proteins/metabolism , Amino Acid Sequence , Conserved Sequence , Cucurbita/metabolism , Molecular Sequence Data , Peptide Initiation Factors/genetics , Plant Proteins/genetics , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Processing, Post-Translational , RNA-Binding Proteins/genetics , Eukaryotic Translation Initiation Factor 5A
12.
Eur J Neurosci ; 31(9): 1606-15, 2010 May.
Article in English | MEDLINE | ID: mdl-20525073

ABSTRACT

Many members of the C1q family, including complement C1q and adiponectin, and the structurally related tumor necrosis factor family are secreted and play crucial roles in intercellular signaling. Among them, the Cbln (precerebellin) and C1q-like (C1ql) subfamilies are highly and predominantly expressed in the central nervous system. Although the Cbln subfamily serve as essential trans-neuronal regulators of synaptic integrity in the cerebellum, the functions of the C1ql subfamily (C1ql1-C1ql4) remain unexplored. Here, we investigated the gene expression of the C1ql subfamily in the adult and developing mouse brain by reverse transcriptase-polymerase chain reaction and high-resolution in-situ hybridization. In the adult brain, C1ql1-C1ql3 mRNAs were mainly expressed in neurons but weak expression was seen in glia-like structures in the adult brain. The C1ql1 mRNA was predominantly expressed in the inferior olive, whereas the C1ql2 and C1ql3 mRNAs were strongly coexpressed in the dentate gyrus. Although the C1ql1 and C1ql3 mRNAs were detectable as early as embryonic day 13, the C1ql2 mRNA was observed at later embryonic stages. The C1ql1 mRNA was also expressed transiently in the external granular layer of the cerebellum. Biochemical characterization in heterologous cells revealed that all of the C1ql subfamily proteins were secreted and they formed both homomeric and heteromeric complexes. They also formed hexameric and higher-order complexes via their N-terminal cysteine residues. These results suggest that, like Cbln, the C1ql subfamily has distinct spatial and temporal expression patterns and may play diverse roles by forming homomeric and heteromeric complexes in the central nervous system.


Subject(s)
Brain/growth & development , Brain/metabolism , Complement C1q/metabolism , Aging , Amino Acid Sequence , Animals , Brain/embryology , Cell Line , Cerebellum/embryology , Cerebellum/growth & development , Cerebellum/metabolism , Complement C1q/genetics , Dentate Gyrus/embryology , Dentate Gyrus/growth & development , Dentate Gyrus/metabolism , Humans , Mice , Neuroglia/metabolism , Neurons/metabolism , Olivary Nucleus/embryology , Olivary Nucleus/growth & development , Olivary Nucleus/metabolism , Protein Multimerization , RNA, Messenger/metabolism , Sequence Homology, Amino Acid
13.
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
14.
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
15.
Eur J Neurosci ; 29(4): 693-706, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19250438

ABSTRACT

Cbln1 (a.k.a. precerebellin) is secreted from cerebellar granule cells as homohexamer or in heteromeric complexes with Cbln3. Cbln1 plays crucial roles in regulating morphological integrity of parallel fiber (PF)-Purkinje cell (PC) synapses and synaptic plasticity. Cbln1-knockout mice display severe cerebellar phenotypes that are essentially indistinguishable from those in glutamate receptor GluRdelta2-null mice, and include severe reduction in the number of PF-PC synapses and loss of long-term depression of synaptic transmission. To understand better the relationship between Cbln1, Cbln3 and GluRdelta2, we performed light and electron microscopic immunohistochemical analyses using highly specific antibodies and antigen-exposing methods, i.e. pepsin pretreatment for light microscopy and postembedding immunogold for electron microscopy. In conventional immunohistochemistry, Cbln1 was preferentially associated with non-terminal portions of PF axons in the molecular layer but rarely overlapped with Cbln3. In contrast, antigen-exposing methods not only greatly intensified Cbln1 immunoreactivity in the molecular layer, but also revealed its high accumulation in the synaptic cleft of PF-PC synapses. No such synaptic accumulation was evident at other PC synapses. Furthermore, Cbln1 now came to overlap almost completely with Cbln3 and GluRdelta2 at PF-PC synapses. Therefore, the convergence of all three molecules provides the anatomical basis for a common signaling pathway regulating circuit development and synaptic plasticity in the cerebellum.


Subject(s)
Cerebellum/metabolism , Nerve Tissue Proteins/metabolism , Protein Precursors/metabolism , Purkinje Cells/metabolism , Receptors, Glutamate/metabolism , Synapses/metabolism , Animals , Fluorescent Antibody Technique , Immunohistochemistry , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Immunoelectron , Nerve Tissue Proteins/genetics , Pepsin A , Protein Precursors/genetics
16.
Nat Neurosci ; 8(11): 1534-41, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16234806

ABSTRACT

Cbln1 is a cerebellum-specific protein of previously unknown function that is structurally related to the C1q and tumor necrosis factor families of proteins. We show that Cbln1 is a glycoprotein secreted from cerebellar granule cells that is essential for three processes in cerebellar Purkinje cells: the matching and maintenance of pre- and postsynaptic elements at parallel fiber-Purkinje cell synapses, the establishment of the proper pattern of climbing fiber-Purkinje cell innervation, and induction of long-term depression at parallel fiber-Purkinje cell synapses. Notably, the phenotype of cbln1-null mice mimics loss-of-function mutations in the orphan glutamate receptor, GluR delta2, a gene selectively expressed in Purkinje neurons. Therefore, Cbln1 secreted from presynaptic granule cells may be a component of a transneuronal signaling pathway that controls synaptic structure and plasticity.


Subject(s)
Cerebellum/cytology , Nerve Tissue Proteins/physiology , Neuronal Plasticity/physiology , Neurons/physiology , Protein Precursors/physiology , Synapses/metabolism , Age Factors , Analysis of Variance , Animals , Animals, Newborn , Ataxia/genetics , Behavior, Animal , Blotting, Northern/methods , Blotting, Western/methods , Cells, Cultured , Cloning, Molecular/methods , Dendritic Spines , Dose-Response Relationship, Radiation , Electric Stimulation/methods , Excitatory Postsynaptic Potentials/physiology , Excitatory Postsynaptic Potentials/radiation effects , Gene Expression Regulation , Humans , In Situ Hybridization/methods , In Vitro Techniques , Membrane Potentials/physiology , Membrane Potentials/radiation effects , Mice , Mice, Inbred ICR , Mice, Transgenic , Microscopy, Electron, Transmission/methods , Motor Activity/genetics , Mutagenesis/physiology , Nerve Tissue Proteins/genetics , Neurons/ultrastructure , Patch-Clamp Techniques/methods , Protein Precursors/genetics , RNA, Messenger/metabolism , Radioimmunoassay/methods , Reverse Transcriptase Polymerase Chain Reaction/methods , Synapses/ultrastructure , Transfection/methods , Vesicular Glutamate Transport Protein 2/metabolism
17.
Clin Nutr ESPEN ; 28: 67-73, 2018 12.
Article in English | MEDLINE | ID: mdl-30390895

ABSTRACT

BACKGROUND & AIMS: Systemic inflammation plays an important role in the pathogenesis of chronic obstructive pulmonary disease (COPD), resulting in depletion of lean body mass (LBM) and muscle mass. Both frequent exacerbation of COPD and low LBM are associated with poor prognosis. This study aimed to evaluate whether supplementation of eicosapentaenoic acid (EPA) prevents depletion of LBM and muscle mass in hospitalized patients with exacerbation of COPD. METHODS: This was a prospective randomized controlled trial, conducted between November 2014 and October 2017. Fifty patients were randomly assigned to receive 1 g/day of EPA-enriched oral nutrition supplementation (ONS) (EPA group) or EPA-free ONS of similar energy (control group) during hospitalization. The LBM index (LBMI) and the skeletal muscle mass index (SMI) were measured using a bioelectrical impedance analyzer at the time of admission and at the time of discharge. Patients underwent pulmonary rehabilitation and wore a pedometer to measure step counts and physical activity. RESULTS: Forty-five patients that completed the experiment were analyzed. Baseline characteristics were similar between the EPA (n = 24) and control groups (n = 21). There were no significant differences in energy intake, step counts, physical activity, or length of hospitalization between the two groups. Although the plasma levels of EPA significantly increased only in the EPA group, we found an insignificant increase in LBMI and SMI in the EPA group compared with the control group (LBMI: +0.35 vs. +0.19 kg/m2, P = 0.60, and SMI: +0.2 vs. -0.3 kg/m2, P = 0.17, respectively). The change in the SMI was significantly correlated with the length of hospitalization in the EPA group, but not in the control group (r = 0.53, P = 0.008, and r = -0.09, P = 0.70, respectively). CONCLUSIONS: EPA-enriched ONS in patients with exacerbation of COPD during short-time hospitalization had no significant advantage in preservation of LBM and muscle mass compared with EPA-free ONS. EPA supplementation for a longer duration might play an important role in the recovery of skeletal muscle mass after exacerbation of COPD.


Subject(s)
Cachexia/prevention & control , Dietary Supplements , Eicosapentaenoic Acid , Pulmonary Disease, Chronic Obstructive , Aged , Body Composition , Female , Humans , Male , Nutritional Status , Prospective Studies , Treatment Outcome
18.
Neuron ; 99(5): 985-998.e6, 2018 09 05.
Article in English | MEDLINE | ID: mdl-30122381

ABSTRACT

Long-term depression (LTD) of AMPA-type glutamate receptor (AMPA receptor)-mediated synaptic transmission has been proposed as a cellular substrate for learning and memory. Although activity-induced AMPA receptor endocytosis is believed to underlie LTD, it remains largely unclear whether LTD and AMPA receptor endocytosis at specific synapses are causally linked to learning and memory in vivo. Here we developed a new optogenetic tool, termed PhotonSABER, which enabled the temporal, spatial, and cell-type-specific control of AMPA receptor endocytosis at active synapses, while the basal synaptic properties and other forms of synaptic plasticity were unaffected. We found that fiberoptic illumination to Purkinje cells expressing PhotonSABER in vivo inhibited cerebellar motor learning during adaptation of the horizontal optokinetic response and vestibulo-ocular reflex, as well as synaptic AMPA receptor decrease in the flocculus. Our results demonstrate that LTD and AMPA receptor endocytosis at specific neuronal circuits were directly responsible for motor learning in vivo. VIDEO ABSTRACT.


Subject(s)
Endocytosis/physiology , Learning/physiology , Long-Term Synaptic Depression/physiology , Motor Activity/physiology , Optogenetics/methods , Receptors, AMPA/physiology , Animals , Cells, Cultured , HEK293 Cells , Humans , Mice , Mice, Inbred C57BL , Mice, Inbred ICR , Organ Culture Techniques , Purkinje Cells/chemistry , Purkinje Cells/physiology , Receptors, AMPA/analysis
19.
Commun Biol ; 1: 96, 2018.
Article in English | MEDLINE | ID: mdl-30175250

ABSTRACT

Mutations in the SCN2A gene encoding a voltage-gated sodium channel Nav1.2 are associated with epilepsies, intellectual disability, and autism. SCN2A gain-of-function mutations cause early-onset severe epilepsies, while loss-of-function mutations cause autism with milder and/or later-onset epilepsies. Here we show that both heterozygous Scn2a-knockout and knock-in mice harboring a patient-derived nonsense mutation exhibit ethosuximide-sensitive absence-like seizures associated with spike-and-wave discharges at adult stages. Unexpectedly, identical seizures are reproduced and even more prominent in mice with heterozygous Scn2a deletion specifically in dorsal-telencephalic (e.g., neocortical and hippocampal) excitatory neurons, but are undetected in mice with selective Scn2a deletion in inhibitory neurons. In adult cerebral cortex of wild-type mice, most Nav1.2 is expressed in excitatory neurons with a steady increase and redistribution from proximal (i.e., axon initial segments) to distal axons. These results indicate a pivotal role of Nav1.2 haplodeficiency in excitatory neurons in epilepsies of patients with SCN2A loss-of-function mutations.

20.
J Neurosci ; 26(18): 4740-51, 2006 May 03.
Article in English | MEDLINE | ID: mdl-16672646

ABSTRACT

2-arachidonoyl-glycerol (2-AG) is an endocannabinoid that is released from postsynaptic neurons, acts retrogradely on presynaptic cannabinoid receptor CB1, and induces short- and long-term suppression of transmitter release. To understand the mechanisms of the 2-AG-mediated retrograde modulation, we investigated subcellular localization of a major 2-AG biosynthetic enzyme, diacylglycerol lipase-alpha (DAGLalpha), by using immunofluorescence and immunoelectron microscopy in the mouse brain. In the cerebellum, DAGLalpha was predominantly expressed in Purkinje cells. DAGLalpha was detected on the dendritic surface and occasionally on the somatic surface, with a distal-to-proximal gradient from spiny branchlets toward somata. DAGLalpha was highly concentrated at the base of spine neck and also accumulated with much lower density on somatodendritic membrane around the spine neck. However, DAGLalpha was excluded from the main body of spine neck and head. In hippocampal pyramidal cells, DAGLalpha was also accumulated in spines. In contrast to the distribution in Purkinje cells, DAGLalpha was distributed in the spine head, neck, or both, whereas somatodendritic membrane was labeled very weakly. These results indicate that DAGLalpha is essentially targeted to postsynaptic spines in cerebellar and hippocampal neurons, but its fine distribution within and around spines is differently regulated between the two neurons. The preferential spine targeting should enable efficient 2-AG production on excitatory synaptic activity and its swift retrograde modulation onto nearby presynaptic terminals expressing CB1. Furthermore, different fine localization within and around spines suggests that the distance between postsynaptic 2-AG production site and presynaptic CB1 is differentially controlled depending on neuron types.


Subject(s)
Arachidonic Acids/metabolism , Cannabinoid Receptor Modulators/metabolism , Dendritic Spines/enzymology , Endocannabinoids , Glycerides/metabolism , Lipoprotein Lipase/metabolism , Presynaptic Terminals/enzymology , Receptor, Cannabinoid, CB1/metabolism , Animals , Animals, Newborn , Antibody Specificity , Blotting, Western/methods , Brain/cytology , Brain/enzymology , Dendritic Spines/ultrastructure , Guinea Pigs , Immunohistochemistry/methods , In Situ Hybridization/methods , Mice , Mice, Inbred C57BL , Microscopy, Immunoelectron/methods , Neurons/cytology , Presynaptic Terminals/ultrastructure , Rabbits , Receptors, Metabotropic Glutamate/metabolism , Vesicular Glutamate Transport Protein 2/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL