Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
3.
Front Cell Neurosci ; 18: 1353542, 2024.
Article in English | MEDLINE | ID: mdl-38469354

ABSTRACT

Introduction: Loss of proteasome function, proteinopathy, and proteotoxicity may cause neurodegeneration across the human lifespan in several forms of brain injury and disease. Drugs that activate brain proteasomes in vivo could thus have a broad therapeutic impact in neurology. Methods: Using pigs, a clinically relevant large animal with a functionally compartmental gyrencephalic cerebral cortex, we evaluated the localization and biochemical activity of brain proteasomes and tested the ability of small molecules to activate brain proteasomes. Results: By Western blotting, proteasome protein subunit PSMB5 and PSMA3 levels were similar in different pig brain regions. Immunohistochemistry for PSMB5 showed localization in the cytoplasm (diffuse and particulate) and nucleus (cytoplasm < nucleus). Some PSMB5 immunoreactivity was colocalized with mitochondrial (voltage-gated anion channel and cyclophilin D) and cell death (Aven) proteins in the neuronal soma and neuropil in the neocortex of pig and human brains. In the nucleus, PSMB5 immunoreactivity was diffuse, particulate, and clustered, including perinucleolar decorations. By fluorogenic assay, proteasome chymotrypsin-like activities (CTL) in crude tissue soluble fractions were generally similar within eight different pig brain regions. Proteasome CTL activity in the hippocampus was correlated with activity in nasal mucosa biopsies. In pilot analyses of subcellular fractions of pig cerebral cortex, proteasome CTL activity was highest in the cytosol and then ~50% lower in nuclear fractions; ~15-20% of total CTL activity was in pure mitochondrial fractions. With in-gel activity assay, 26S-singly and -doubly capped proteasomes were the dominant forms in the pig cerebral cortex. With a novel in situ histochemical activity assay, MG132-inhibitable proteasome CTL activity was localized to the neuropil, as a mosaic, and to cell bodies, nuclei, and centrosome-like perinuclear satellites. In piglets treated intravenously with pyrazolone derivative and chlorpromazine over 24 h, brain proteasome CTL activity was modestly increased. Discussion: This study shows that the proteasome in the pig brain has relative regional uniformity, prominent nuclear and perinuclear presence with catalytic activity, a mitochondrial association with activity, 26S-single cap dominance, and indications from small molecule systemic administration of pyrazolone derivative and chlorpromazine that brain proteasome function appears safely activable.

4.
BMB Rep ; 55(5): 232-237, 2022 May.
Article in English | MEDLINE | ID: mdl-35410636

ABSTRACT

The Wnt/ß-catenin signaling plays crucial roles in early development, tissue homeostasis, stem cells, and cancers. Here, we show that RNF152, an E3 ligase localized to lysosomes, acts as a negative regulator of the Wnt/ß-catenin pathway during Xenopus early embryogenesis. Overexpression of wild-type (WT) RNF152 inhibited XWnt8-induced stabilization of ß-catenin, ectopic expression of target genes, and activity of a Wnt-responsive promoter. Likewise, an E3 ligase-defective RNF152 had repressive effects on the Wnt-dependent gene responses but not its truncation mutant lacking the transmembrane domain. Conversely, knockdown of RNF152 further enhanced the transcriptional responses induced by XWnt8. RNF152 morphants exhibited defects in craniofacial structures and pigmentation. In line with this, the gain-of-RNF152 function interfered with the expression of neural crest (NC) markers, whereas its depletion up-regulated NC formation in the early embryo. Mechanistically, RNF152 inhibits the polymerization of Dishevelled, which is key to Wnt signaling, in an E3 ligase-independent manner. Together, these results suggest that RNF152 controls negatively Wnt/ß-catenin signaling to fine-tune its activity for NC formation in Xenopus embryo. [BMB Reports 2022; 55(5): 232-237].


Subject(s)
Wnt Signaling Pathway , beta Catenin , Animals , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Xenopus Proteins/genetics , Xenopus Proteins/metabolism , Xenopus laevis , beta Catenin/metabolism
5.
Biol Psychiatry ; 91(9): 821-831, 2022 05 01.
Article in English | MEDLINE | ID: mdl-35219498

ABSTRACT

BACKGROUND: IQSEC3, a gephyrin-binding GABAergic (gamma-aminobutyric acidergic) synapse-specific guanine nucleotide exchange factor, was recently reported to regulate activity-dependent GABAergic synapse maturation, but the underlying signaling mechanisms remain incompletely understood. METHODS: We generated mice with conditional knockout (cKO) of Iqsec3 to examine whether altered synaptic inhibition influences hippocampus-dependent fear memory formation. In addition, electrophysiological recordings, immunohistochemistry, and behavioral assays were used to address our question. RESULTS: We found that Iqsec3-cKO induces a specific reduction in GABAergic synapse density, GABAergic synaptic transmission, and maintenance of long-term potentiation in the hippocampal CA1 region. In addition, Iqsec3-cKO mice exhibited impaired fear memory formation. Strikingly, Iqsec3-cKO caused abnormally enhanced activation of ribosomal P70-S6K1-mediated signaling in the hippocampus but not in the cortex. Furthermore, inhibiting upregulated S6K1 signaling by expressing dominant-negative S6K1 in the hippocampal CA1 of Iqsec3-cKO mice completely rescued impaired fear learning and inhibitory synapse density but not deficits in long-term potentiation maintenance. Finally, upregulated S6K1 signaling was rescued by IQSEC3 wild-type, but not by an ARF-GEF (adenosine diphosphate ribosylation factor-guanine nucleotide exchange factor) inactive IQSEC3 mutant. CONCLUSIONS: Our results suggest that IQSEC3-mediated balanced synaptic inhibition in hippocampal CA1 is critical for the proper formation of hippocampus-dependent fear memory.


Subject(s)
Fear , Guanine Nucleotide Exchange Factors , Hippocampus , Synapses , Animals , Guanine Nucleotide Exchange Factors/genetics , Guanine Nucleotide Exchange Factors/metabolism , Hippocampus/metabolism , Long-Term Potentiation , Mice , Mice, Inbred C57BL , Mice, Knockout , Synapses/metabolism , Up-Regulation
6.
Proc Natl Acad Sci U S A ; 119(3)2022 01 18.
Article in English | MEDLINE | ID: mdl-35022233

ABSTRACT

Synaptic cell-adhesion molecules (CAMs) organize the architecture and properties of neural circuits. However, whether synaptic CAMs are involved in activity-dependent remodeling of specific neural circuits is incompletely understood. Leucine-rich repeat transmembrane protein 3 (LRRTM3) is required for the excitatory synapse development of hippocampal dentate gyrus (DG) granule neurons. Here, we report that Lrrtm3-deficient mice exhibit selective reductions in excitatory synapse density and synaptic strength in projections involving the medial entorhinal cortex (MEC) and DG granule neurons, accompanied by increased neurotransmitter release and decreased excitability of granule neurons. LRRTM3 deletion significantly reduced excitatory synaptic innervation of hippocampal mossy fibers (Mf) of DG granule neurons onto thorny excrescences in hippocampal CA3 neurons. Moreover, LRRTM3 loss in DG neurons significantly decreased mossy fiber long-term potentiation (Mf-LTP). Remarkably, silencing MEC-DG circuits protected against the decrease in the excitatory synaptic inputs onto DG and CA3 neurons, excitability of DG granule neurons, and Mf-LTP in Lrrtm3-deficient mice. These results suggest that LRRTM3 may be a critical factor in activity-dependent synchronization of the topography of MEC-DG-CA3 excitatory synaptic connections. Collectively, our data propose that LRRTM3 shapes the target-specific structural and functional properties of specific hippocampal circuits.


Subject(s)
Cortical Synchronization/physiology , Hippocampus/physiology , Membrane Proteins/metabolism , Nerve Net/physiology , Nerve Tissue Proteins/metabolism , Synapses/physiology , Animals , CA3 Region, Hippocampal/metabolism , Dentate Gyrus/metabolism , Entorhinal Cortex/metabolism , Long-Term Potentiation , Membrane Proteins/deficiency , Mice, Knockout , Mossy Fibers, Hippocampal/metabolism , Nerve Tissue Proteins/deficiency , Neurons/metabolism , Pseudopodia/metabolism , Synaptic Transmission/physiology
7.
Cell Rep ; 36(3): 109417, 2021 07 20.
Article in English | MEDLINE | ID: mdl-34289353

ABSTRACT

Activity-dependent GABAergic synapse plasticity is important for normal brain functions, but the underlying molecular mechanisms remain incompletely understood. Here, we show that Npas4 (neuronal PAS-domain protein 4) transcriptionally regulates the expression of IQSEC3, a GABAergic synapse-specific guanine nucleotide-exchange factor for ADP-ribosylation factor (ARF-GEF) that directly interacts with gephyrin. Neuronal activation by an enriched environment induces Npas4-mediated upregulation of IQSEC3 protein specifically in CA1 stratum oriens layer somatostatin (SST)-expressing GABAergic interneurons. SST+ interneuron-specific knockout (KO) of Npas4 compromises synaptic transmission in these GABAergic interneurons, increases neuronal activity in CA1 pyramidal neurons, and reduces anxiety behavior, all of which are normalized by the expression of wild-type IQSEC3, but not a dominant-negative ARF-GEF-inactive mutant, in SST+ interneurons of Npas4-KO mice. Our results suggest that IQSEC3 is a key GABAergic synapse component that is directed by Npas4 and ARF activity, specifically in SST+ interneurons, to orchestrate excitation-to-inhibition balance and control anxiety-like behavior.


Subject(s)
Anxiety/metabolism , Basic Helix-Loop-Helix Transcription Factors/metabolism , Behavior, Animal , Guanine Nucleotide Exchange Factors/metabolism , Hippocampus/metabolism , Interneurons/metabolism , Somatostatin/metabolism , Animals , GABAergic Neurons/metabolism , Mice, Inbred C57BL , Mice, Knockout , Promoter Regions, Genetic/genetics , Protein Binding , Synapses/metabolism , Synaptic Transmission , Up-Regulation
8.
iScience ; 24(2): 102037, 2021 Feb 19.
Article in English | MEDLINE | ID: mdl-33532714

ABSTRACT

Gephyrin is critical for the structure, function, and plasticity of inhibitory synapses. Gephyrin mutations have been linked to various neurological disorders; however, systematic analyses of the functional consequences of these mutations are lacking. Here, we performed molecular dynamics simulations of gephyrin to predict how six reported point mutations might change the structural stability and/or function of gephyrin. Additional in silico analyses revealed that the A91T and G375D mutations reduce the binding free energy of gephyrin oligomer formation. Gephyrin A91T and G375D displayed altered clustering patterns in COS-7 cells and nullified the inhibitory synapse-promoting effect of gephyrin in cultured neurons. However, only the G375D mutation reduced gephyrin interaction with GABAA receptors and neuroligin-2 in mouse brain; it also failed to normalize deficits in GABAergic synapse maintenance and neuronal hyperactivity observed in hippocampal dentate gyrus-specific gephyrin-deficient mice. Our results provide insights into biochemical, cell-biological, and network-activity effects of the pathogenic G375D mutation.

9.
Glia ; 68(12): 2661-2673, 2020 12.
Article in English | MEDLINE | ID: mdl-32645240

ABSTRACT

IQSEC3, a guanine nucleotide exchange factor for ADP-ribosylation factors (ARF-GEFs) is specifically expressed at GABAergic synapses, and its loss increases seizure susceptibility in mice. However, the contribution of microglia to initiation and/or progression of seizures in IQSEC3-deficient mice has not been investigated. In the current study, we show that mice with hippocampal dentate gyrus (DG)-specific IQSEC3 knockdown (KD) exhibit microglial activation and death of DG granule cell. Furthermore, treatment of IQSEC3-KD mice with minocycline, an inhibitor of microglial activation, blocks DG granule neuron cell death and the occurrence of spontaneous seizures without affecting GABAergic synapse deficits or loss of somatostatin. Our results suggest that microglial activation is involved in a subset of IQSEC3-KD-induced epileptogenesis stages, and that its regulation could be an alternative strategy for managing epilepsy.


Subject(s)
Microglia , Seizures , Animals , Dentate Gyrus , Guanine Nucleotide Exchange Factors , Hippocampus , Mice , Neurons , Synapses
10.
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
11.
EMBO Rep ; 21(5): e48693, 2020 05 06.
Article in English | MEDLINE | ID: mdl-32103600

ABSTRACT

The tumor suppressor Smad4, a key mediator of the TGF-ß/BMP pathways, is essential for development and tissue homeostasis. Phosphorylation of Smad4 in its linker region catalyzed by the mitogen-activated protein kinase (MAPK) plays a pivotal role in regulating its transcriptional activity and stability. In contrast, roles of Smad4 dephosphorylation as a control mechanism of TGF-ß/BMP signaling and the phosphatases responsible for its dephosphorylation remain so far elusive. Here, we identify Wip1 as a Smad4 phosphatase. Wip1 selectively binds and dephosphorylates Smad4 at Thr277, a key MAPK phosphorylation site, thereby regulating its nuclear accumulation and half-life. In Xenopus embryos, Wip1 limits mesoderm formation and favors neural induction by inhibiting TGF-ß/BMP signals. Wip1 restrains TGF-ß-induced growth arrest, migration, and invasion in human cells and enhances the tumorigenicity of cancer cells by repressing the antimitogenic activity of Smad4. We propose that Wip1-dependent dephosphorylation of Smad4 is critical for the regulation of TGF-ß signaling.


Subject(s)
Protein Phosphatase 2C/metabolism , Signal Transduction , Smad4 Protein/metabolism , Transforming Growth Factor beta , Xenopus Proteins/metabolism , Animals , Humans , Mitogen-Activated Protein Kinases/metabolism , Phosphorylation , Protein Phosphatase 2C/genetics , Smad4 Protein/genetics , Transforming Growth Factor beta/metabolism , Xenopus Proteins/genetics , Xenopus laevis/metabolism
12.
Cell Rep ; 30(6): 1995-2005.e5, 2020 02 11.
Article in English | MEDLINE | ID: mdl-32049026

ABSTRACT

Gephyrin interacts with various GABAergic synaptic proteins to organize GABAergic synapse development. Among the multitude of gephyrin-binding proteins is IQSEC3, a recently identified component at GABAergic synapses that acts through its ADP ribosylation factor-guanine nucleotide exchange factor (ARF-GEF) activity to orchestrate GABAergic synapse formation. Here, we show that IQSEC3 knockdown (KD) reduced GABAergic synaptic density in vivo, suggesting that IQSEC3 is required for GABAergic synapse maintenance in vivo. We further show that IQSEC3 KD in the dentate gyrus (DG) increases seizure susceptibility and triggers selective depletion of somatostatin (SST) peptides in the DG hilus in an ARF-GEP activity-dependent manner. Strikingly, selective introduction of SST into SST interneurons in DG-specific IQSEC3-KD mice reverses GABAergic synaptic deficits. Thus, our data suggest that IQSEC3 is required for linking gephyrin-GABAA receptor complexes with ARF-dependent pathways to prevent aberrant, runaway excitation and thereby contributes to the integrity of SST interneurons and proper GABAergic synapse maintenance.


Subject(s)
Guanine Nucleotide Exchange Factors/deficiency , Guanine Nucleotide Exchange Factors/metabolism , Hippocampus/metabolism , Membrane Proteins/metabolism , Somatostatin/metabolism , Synapses/metabolism , Animals , HEK293 Cells , Hippocampus/cytology , Humans , Male , Mice , Mice, Inbred C57BL
13.
BMB Rep ; 51(12): 636-641, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30463640

ABSTRACT

DPP4 (dipeptidyl peptidase-4), a highly conserved transmembrane glycoprotein with an exo-peptidase activity, has been shown to contribute to glucose metabolism, immune regulation, signal transduction, and cell differentiation. Here, we show that DPP4 is involved in control of activin/nodal signaling in Xenopus early development. In support of this, gain of function of DPP4 augmented Smad2 phosphorylation as well as expression of target genes induced by activin or nodal signal. In addition, Dpp4 and Xnr1 showed synergistic effect on induction of ectopic dorsal body axis, when co-injected at suboptimal doses in early embryos. Conversely, saxagliptin, a DPP4 inhibitor repressed activin induction of Smad2 phosphorylation. Notably, overexpression of Dpp4 disrupted specification of dorsal body axis of embryo, leading to malformed phenotypes such as spina bifida and a shortened and dorsally bent axis. Together, these results suggest that DPP4 functions as a potentiator of activin/nodal signaling pathway. [BMB Reports 2018; 51(12): 636-641].


Subject(s)
Activins/metabolism , Dipeptidyl Peptidase 4/metabolism , Xenopus Proteins/metabolism , Adamantane/analogs & derivatives , Adamantane/pharmacology , Animals , Dipeptides/pharmacology , Dipeptidyl Peptidase 4/chemistry , Dipeptidyl Peptidase 4/genetics , Dipeptidyl-Peptidase IV Inhibitors/pharmacology , Embryo, Nonmammalian/metabolism , Embryonic Development , HEK293 Cells , Humans , Phosphorylation , Signal Transduction/drug effects , Smad2 Protein/metabolism , Xenopus/growth & development , Xenopus/metabolism
14.
Mol Cells ; 41(5): 373-380, 2018 May 31.
Article in English | MEDLINE | ID: mdl-29665671

ABSTRACT

Synapses and neural circuits form with exquisite specificity during brain development to allow the precise and appropriate flow of neural information. Although this property of synapses and neural circuits has been extensively investigated for more than a century, molecular mechanisms underlying this property are only recently being unveiled. Recent studies highlight several classes of cell-surface proteins as organizing hubs in building structural and functional architectures of specific synapses and neural circuits. In the present mini-review, we discuss recent findings on various synapse organizers that confer the distinct properties of specific synapse types and neural circuit architectures in mammalian brains, with a particular focus on the hippocampus and cerebellum.


Subject(s)
Cerebellum/physiology , Hippocampus/physiology , Neural Cell Adhesion Molecules/physiology , Synapses/physiology , Synaptic Transmission/physiology , Animals , Cell Adhesion , Cerebellum/cytology , Entorhinal Cortex/cytology , Entorhinal Cortex/physiology , Hippocampus/cytology , Humans , Interneurons/physiology , Nerve Tissue Proteins/physiology , Neural Pathways/physiology , Neurons/physiology , Purkinje Cells/physiology
15.
Mol Cells ; 40(11): 823-827, 2017 Nov 30.
Article in English | MEDLINE | ID: mdl-29179261

ABSTRACT

Genome editing using programmable nucleases such as CRISPR/Cas9 or Cpf1 has emerged as powerful tools for gene knock-out or knock-in in various organisms. While most genetic diseases are caused by point mutations, these genome-editing approaches are inefficient in inducing single-nucleotide substitutions. Recently, Cas9-linked cytidine deaminases, named base editors (BEs), have been shown to convert cytidine to uridine efficiently, leading to targeted single-base pair substitutions in human cells and organisms. Here, we first report on the generation of Xenopus laevis mutants with targeted single-base pair substitutions using this RNA-guided programmable deaminase. Injection of base editor 3 (BE3) ribonucleoprotein targeting the tyrosinase (tyr) gene in early embryos can induce site-specific base conversions with the rates of up to 20.5%, resulting in oculocutaneous albinism phenotypes without off-target mutations. We further test this base-editing system by targeting the tp53 gene with the result that the expected single-base pair substitutions are observed at the target site. Collectively, these data establish that the programmable deaminases are efficient tools for creating targeted point mutations for human disease modeling in Xenopus.


Subject(s)
Albinism, Oculocutaneous/genetics , Cytidine Deaminase/metabolism , Monophenol Monooxygenase/genetics , RNA, Guide, Kinetoplastida/genetics , Xenopus laevis/embryology , Amino Acid Substitution , Animals , Gene Editing/methods , Mutation Rate , Phenotype , Point Mutation , Xenopus Proteins/genetics , Xenopus laevis/genetics
16.
Neuron ; 94(6): 1121-1131.e6, 2017 Jun 21.
Article in English | MEDLINE | ID: mdl-28641111

ABSTRACT

Membrane-associated mucin domain-containing glycosylphosphatidylinositol anchor proteins (MDGAs) bind directly to neuroligin-1 (NL1) and neuroligin-2 (NL2), thereby respectively regulating excitatory and inhibitory synapse development. However, the mechanisms by which MDGAs modulate NL activity to specify development of the two synapse types remain unclear. Here, we determined the crystal structures of human NL2/MDGA1 Ig1-3 complex, revealing their stable 2:2 arrangement with three interaction interfaces. Cell-based assays using structure-guided, site-directed MDGA1 mutants showed that all three contact patches were required for the MDGA's negative regulation of NL2-mediated synaptogenic activity. Furthermore, MDGA1 competed with neurexins for NL2 via its Ig1 domain. The binding affinities of both MDGA1 and MDGA2 for NL1 and NL2 were similar, consistent with the structural prediction of similar binding interfaces. However, MDGA1 selectively associated with NL2, but not NL1, in vivo. These findings collectively provide structural insights into the mechanism by which MDGAs negatively modulate synapse development governed by NLs/neurexins.


Subject(s)
Cell Adhesion Molecules, Neuronal/metabolism , Cell Adhesion , GPI-Linked Proteins/metabolism , Nerve Tissue Proteins/metabolism , Neural Cell Adhesion Molecules/metabolism , Protein Binding , Synapses/metabolism , Animals , COS Cells , Calcium-Binding Proteins , Chlorocebus aethiops , Crystallography , HEK293 Cells , Humans , L Cells , Mass Spectrometry , Mice , Neural Inhibition , Neurogenesis , Protein Structure, Quaternary
17.
Dev Biol ; 421(2): 183-193, 2017 Jan 15.
Article in English | MEDLINE | ID: mdl-27913219

ABSTRACT

During early embryogenesis, FGF signals regulate the antero-posterior (AP) patterning of the neural plate by promoting posterior cell fates. In particular, BMP signal-mediated attenuation of FGF pathway plays a critical role in the determination of the anterior neural region. Here we show that Tbx2, a T-box transcriptional repressor regulates anterior neural specification by suppressing FGF8 signaling pathway in Xenopus embryo. Tbx2 is expressed in the anterior edge of the neural plate in early neurulae. Overexpression and knockdown of Tbx2 induce expansion and reduction in the expression of anterior neural markers, respectively. It also suppresses FGF8-induced ERK phosphorylation and neural caudalization. Tbx2, which is a target gene of BMP signal, down-regulates FGF8 signaling by inhibiting the expression of Flrt3, a positive regulator of this pathway. We found that Tbx2 binds directly to the T-box element located in the promoter region of Flrt3 gene, thereby interfering with the activity of the promoter. Consistently, Tbx2 augmentation of anterior neural formation is inhibited by co-expression of Flrt3. Furthermore, disruption of the anterior-most structures such as eyes in Tbx2-depleted embryos can be rescued by inhibition of Flrt3 function or FGF signaling. Taken together, our results suggest that Tbx2 mediates BMP signal to down-regulate FGF signaling pathway by repressing Flrt3 expression for anterior tissue formation.


Subject(s)
Body Patterning/genetics , Fibroblast Growth Factors/metabolism , Nervous System/embryology , Nervous System/metabolism , Signal Transduction , T-Box Domain Proteins/metabolism , Xenopus Proteins/metabolism , Xenopus laevis/embryology , Animals , Base Sequence , Bone Morphogenetic Proteins/metabolism , Brain/embryology , Brain/metabolism , Down-Regulation/genetics , Gene Expression Regulation, Developmental , Head/embryology , In Situ Hybridization , Membrane Proteins/genetics , Membrane Proteins/metabolism , Models, Biological , Promoter Regions, Genetic/genetics , T-Box Domain Proteins/genetics , Xenopus Proteins/genetics , Xenopus laevis/genetics
18.
Biochem Biophys Res Commun ; 478(1): 455-461, 2016 09 09.
Article in English | MEDLINE | ID: mdl-27318088

ABSTRACT

Xenopus embryo serves as an ideal model for teratogenesis assays to examine the effects of any substances on the cellular processes critical for early development and adult tissue homeostasis. In our chemical library screening with frog embryo, capsaicin was found to repress the Wnt/ß-catenin signaling. Depending on the stages at which embryos became exposed to capsaicin, it could disrupt formation of dorsal or posterior body axis of embryo, which is associated with inhibition of maternal or zygotic Wnt signal in early development. In agreement with these phenotypes, capsaicin suppressed the expression of Wnt target genes such as Siamois and Chordin in the organizer region of embryo and in Wnt signals-stimulated tissue explants. In addition, the cellular level of ß-catenin, a key component of Wnt pathway, was down-regulated in capsaicin-treated embryonic cells. Unlike wild-type ß-catenin, its non-phosphorylatable mutant in which serine and threonine residues phosphorylated by GSK3 are substituted with alanine was not destabilized by capsaicin, indicative of the effect of this chemical on the phosphorylation status of ß-catenin. In support of this, capsaicin up-regulated the level of GSK3- or CK1-phosphorylated ß-catenin, concomitantly lowering that of its de-phosphorylated version. Notably, capsaicin augmented the phosphorylation of a phosphatase, PP2A at tyrosine 307, suggesting its repression of the enzymatic activity of the phosphatase. Furthermore, capsaicin still enhanced ß-catenin phosphorylation in cells treated with a GSK3 inhibitor, LiCl but not in those treated with a phosphatase inhibitor, okadaic acid. Together, these results indicate that capsaicin inhibits the patterning of the dorso-ventral and anterior-posterior body axes of embryo by repressing PP2A and thereby down-regulating the Wnt/ß-catenin signaling.


Subject(s)
Body Patterning/drug effects , Capsaicin/toxicity , Down-Regulation/drug effects , Protein Phosphatase 2/metabolism , Teratogens/toxicity , Wnt Signaling Pathway/drug effects , Animals , Dose-Response Relationship, Drug , Gene Expression Regulation, Developmental/drug effects , Xenopus laevis
19.
J Biol Chem ; 291(19): 10119-30, 2016 May 06.
Article in English | MEDLINE | ID: mdl-27002143

ABSTRACT

Gephyrin is a central scaffold protein that mediates development, function, and plasticity of mammalian inhibitory synapses by interacting with various inhibitory synaptic proteins. Here, we show that IQSEC3, a guanine nucleotide exchange factor for ARF6, directly interacts with gephyrin, an interaction that is critical for the inhibitory synapse localization of IQSEC3. Overexpression of IQSEC3 increases inhibitory, but not excitatory, synapse density in a guanine nucleotide exchange factor activity-dependent manner. Conversely, knockdown of IQSEC3 decreases size of gephyrin cluster without altering gephyrin puncta density. Collectively, these data reveal that IQSEC3 acts together with gephyrin to regulate inhibitory synapse development.


Subject(s)
Carrier Proteins , Guanine Nucleotide Exchange Factors , Membrane Proteins , Synapses , Animals , Carrier Proteins/genetics , Carrier Proteins/metabolism , Guanine Nucleotide Exchange Factors/genetics , Guanine Nucleotide Exchange Factors/metabolism , Humans , Membrane Proteins/genetics , Membrane Proteins/metabolism , Rats , Synapses/genetics , Synapses/metabolism
20.
Cell Rep ; 14(4): 808-822, 2016 Feb 02.
Article in English | MEDLINE | ID: mdl-26776509

ABSTRACT

The four members of the LRRTM family (LRRTM1-4) are postsynaptic adhesion molecules essential for excitatory synapse development. They have also been implicated in neuropsychiatric diseases. Here, we focus on LRRTM3, showing that two distinct LRRTM3 variants generated by alternative splicing regulate LRRTM3 interaction with PSD-95, but not its excitatory synapse-promoting activity. Overexpression of either LRRTM3 variant increased excitatory synapse density in dentate gyrus (DG) granule neurons, whereas LRRTM3 knockdown decreased it. LRRTM3 also controlled activity-regulated AMPA receptor surface expression in an alternative splicing-dependent manner. Furthermore, Lrrtm3-knockout mice displayed specific alterations in excitatory synapse density, excitatory synaptic transmission and excitability in DG granule neurons but not in CA1 pyramidal neurons. Lastly, LRRTM3 required only specific splice variants of presynaptic neurexins for their synaptogenic activity. Collectively, our data highlight alternative splicing and differential presynaptic ligand utilization in the regulation of LRRTMs, revealing key regulatory mechanisms for excitatory synapse development.


Subject(s)
Alternative Splicing , Cell Adhesion Molecules, Neuronal/metabolism , Excitatory Postsynaptic Potentials , Animals , CA1 Region, Hippocampal/cytology , CA1 Region, Hippocampal/growth & development , CA1 Region, Hippocampal/metabolism , Cell Adhesion Molecules, Neuronal/genetics , Cells, Cultured , Dentate Gyrus/cytology , Dentate Gyrus/growth & development , Dentate Gyrus/metabolism , HEK293 Cells , Humans , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neurogenesis , Protein Transport , Pyramidal Cells/metabolism , Pyramidal Cells/physiology , Rats , Receptors, AMPA/metabolism , Synapses/metabolism , Synapses/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...