Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 158
Filter
Add more filters

Country/Region as subject
Publication year range
1.
J Biol Chem ; 300(2): 105629, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38199563

ABSTRACT

In contrast to stage-specific transcription factors, the role of ubiquitous transcription factors in neuronal development remains a matter of scrutiny. Here, we demonstrated that a ubiquitous factor NF-Y is essential for neural progenitor maintenance during brain morphogenesis. Deletion of the NF-YA subunit in neural progenitors by using nestin-cre transgene in mice resulted in significant abnormalities in brain morphology, including a thinner cerebral cortex and loss of striatum during embryogenesis. Detailed analyses revealed a progressive decline in multiple neural progenitors in the cerebral cortex and ganglionic eminences, accompanied by induced apoptotic cell death and reduced cell proliferation. In neural progenitors, the NF-YA short isoform lacking exon 3 is dominant and co-expressed with cell cycle genes. ChIP-seq analysis from the cortex during early corticogenesis revealed preferential binding of NF-Y to the cell cycle genes, some of which were confirmed to be downregulated following NF-YA deletion. Notably, the NF-YA short isoform disappears and is replaced by its long isoform during neuronal differentiation. Forced expression of the NF-YA long isoform in neural progenitors resulted in a significant decline in neuronal count, possibly due to the suppression of cell proliferation. Collectively, we elucidated a critical role of the NF-YA short isoform in maintaining neural progenitors, possibly by regulating cell proliferation and apoptosis. Moreover, we identified an isoform switch in NF-YA within the neuronal lineage in vivo, which may explain the stage-specific role of NF-Y during neuronal development.


Subject(s)
CCAAT-Binding Factor , Cerebral Cortex , Animals , Mice , CCAAT-Binding Factor/genetics , CCAAT-Binding Factor/metabolism , Cerebral Cortex/cytology , Cerebral Cortex/growth & development , Cerebral Cortex/metabolism , Gene Expression Regulation , Neurogenesis , Protein Isoforms/genetics , Protein Isoforms/metabolism , Transcription Factors/metabolism
2.
Transgenic Res ; 33(3): 99-117, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38684589

ABSTRACT

Golli-myelin basic proteins, encoded by the myelin basic protein gene, are widely expressed in neurons and oligodendrocytes in the central nervous system. Further, prior research has shown that Golli-myelin basic protein is necessary for myelination and neuronal maturation during central nervous system development. In this study, we established Golli-myelin basic protein-floxed mice to elucidate the cell-type-specific effects of Golli-myelin basic protein knockout through the generation of conditional knockout mice (Golli-myelin basic proteinsfl/fl; E3CreN), in which Golli-myelin basic proteins were specifically deleted in cerebellar granule neurons, where Golli-myelin basic proteins are expressed abundantly in wild-type mice. To investigate the role of Golli-myelin basic proteins in cerebellar granule neurons, we further performed histopathological analyses of these mice, with results indicating no morphological changes or degeneration of the major cellular components of the cerebellum. Furthermore, behavioral analysis showed that Golli-myelin basic proteinsfl/fl; E3CreN mice were healthy and did not display any abnormal behavior. These results suggest that the loss of Golli-myelin basic proteins in cerebellar granule neurons does not lead to cerebellar perturbations or behavioral abnormalities. This mouse model could therefore be employed to analyze the effect of Golli-myelin basic protein deletion in specific cell types of the central nervous system, such as other neuronal cells and oligodendrocytes, or in lymphocytes of the immune system.


Subject(s)
Cerebellum , Mice, Knockout , Myelin Basic Protein , Neurons , Animals , Neurons/metabolism , Mice , Cerebellum/metabolism , Cerebellum/growth & development , Myelin Basic Protein/genetics , Myelin Basic Protein/metabolism
3.
J Biol Chem ; 295(29): 9768-9785, 2020 07 17.
Article in English | MEDLINE | ID: mdl-32499373

ABSTRACT

Huntington disease (HD) is a neurodegenerative disorder caused by expanded CAG repeats in the Huntingtin gene. Results from previous studies have suggested that transcriptional dysregulation is one of the key mechanisms underlying striatal medium spiny neuron (MSN) degeneration in HD. However, some of the critical genes involved in HD etiology or pathology could be masked in a common expression profiling assay because of contamination with non-MSN cells. To gain insight into the MSN-specific gene expression changes in presymptomatic R6/2 mice, a common HD mouse model, here we used a transgenic fluorescent protein marker of MSNs for purification via FACS before profiling gene expression with gene microarrays and compared the results of this "FACS-array" with those obtained with homogenized striatal samples (STR-array). We identified hundreds of differentially expressed genes (DEGs) and enhanced detection of MSN-specific DEGs by comparing the results of the FACS-array with those of the STR-array. The gene sets obtained included genes ubiquitously expressed in both MSNs and non-MSN cells of the brain and associated with transcriptional regulation and DNA damage responses. We proposed that the comparative gene expression approach using the FACS-array may be useful for uncovering the gene cascades affected in MSNs during HD pathogenesis.


Subject(s)
Corpus Striatum/metabolism , Flow Cytometry , Huntington Disease/metabolism , Transcriptome , Animals , Corpus Striatum/pathology , Disease Models, Animal , Huntingtin Protein/genetics , Huntingtin Protein/metabolism , Huntington Disease/genetics , Huntington Disease/pathology , Male , Mice , Mice, Transgenic
4.
Biochem Biophys Res Commun ; 522(3): 655-661, 2020 02 12.
Article in English | MEDLINE | ID: mdl-31785806

ABSTRACT

Many pathological proteins related to neurodegenerative diseases are misfolded, aggregating to form amyloid fibrils during pathogenesis. One of the pathological proteins, alpha-synuclein (α-syn), accumulates in the brains of Parkinson disease (PD), dementia with Lewy bodies (DLB) and multiple system atrophy (MSA), which are designated as synucleinopathies. Recently, structural properties of abnormal accumulated proteins are suggested to determine the disease phenotype. However, the biochemical and structural characteristics of those accumulated proteins are still poorly understood. We previously reported the sequence and seed-structure-dependent polymorphic fibrils of α-syn and the polymorphism was identified by proteinase K-resistant cores determined by mass spectrometry (MS) analysis. In this study, we applied this method to analyze α-syn aggregates of MSA and DLB. To perform MS analysis on proteinase K-resistant cores, we first performed amplification of α-syn aggregates by seeding reaction and protein misfolding cyclic amplification (PMCA) to obtain a sufficient amount of aggregates. Using SDS insoluble fraction of the disease brain, we successfully amplified enough α-syn aggregates for MS analysis. We differentiated between mouse and human α-syn aggregates by MS analysis on proteinase K-resistant cores of the aggregates before and after amplification. The results suggest that structural properties of amplified α-syn fibrils are preserved after PMCA and these methods can be applicable in the study of pathological proteins of the neurodegenerative disorders.


Subject(s)
Endopeptidase K/metabolism , Protein Aggregation, Pathological/metabolism , Synucleinopathies/metabolism , alpha-Synuclein/metabolism , Aged , Animals , Brain/metabolism , Brain/pathology , Female , Humans , Male , Mice , Middle Aged , Protein Aggregates , Protein Aggregation, Pathological/pathology , Synucleinopathies/pathology
5.
Biochem Biophys Res Commun ; 508(3): 729-734, 2019 01 15.
Article in English | MEDLINE | ID: mdl-30528390

ABSTRACT

Alpha-synuclein (a-syn) aggregation in brain is implicated in several synucleinopathies, including Parkinson's disease (PD), dementia with Lewy bodies (DLB), and multiple system atrophy (MSA). Until date, at least six disease-associated mutations in a-syn (namely A30P, E46K, H50Q, G51D, A53T, and A53E) are known to cause dominantly inherited familial forms of synucleinopathies. Previous studies using recombinant proteins have reported that a subset of disease-associated mutants show higher aggregation propensities and form spectroscopically distinguishable aggregates compared to wild-type (WT). However, morphological and biochemical comparison of the aggregates for all disease-associated a-syn mutants have not yet been performed. In this study, we performed electron microscopic examination, guanidinium hydrochloride (GdnHCl) denaturation, and protease digestion to classify the aggregates from their respective point mutations. Using electron microscopy we observed variations of amyloid fibrillar morphologies among the aggregates of a-syn mutants, mainly categorized into two groups: twisted fibrils observed for both WT and E46K while straight fibrils for the other mutants. GdnHCl denaturation experiments revealed the a-syn mutants except for E46K were more resistant than WT against the denaturation. Mass spectrometry analysis of protease-treated aggregates showed a variety of protease-resistant cores, which may correspond to their morphological properties. The difference of their properties could be implicated in the clinicopathological difference of synucleinopathies with those mutations.


Subject(s)
Mutant Proteins/metabolism , Parkinson Disease/metabolism , Protein Aggregates , alpha-Synuclein/metabolism , Animals , Endopeptidase K/metabolism , Humans , Mice , Mutant Proteins/chemistry , Mutant Proteins/ultrastructure , Mutation/genetics , alpha-Synuclein/chemistry , alpha-Synuclein/ultrastructure
6.
Mol Cell ; 44(2): 279-89, 2011 Oct 21.
Article in English | MEDLINE | ID: mdl-22017874

ABSTRACT

Selective macroautophagy (autophagy) of ubiquitinated protein is implicated as a compensatory mechanism of the ubiquitin-proteasome system. p62/SQSTM1 is a key molecule managing autophagic clearance of polyubiquitinated proteins. However, little is known about mechanisms controlling autophagic degradation of polyubiquitinated proteins. Here, we show that the specific phosphorylation of p62 at serine 403 (S403) in its ubiquitin-associated (UBA) domain increases the affinity between UBA and polyubiquitin chain, resulting in efficiently targeting polyubiquitinated proteins in "sequestosomes" and stabilizing sequestosome structure as a cargo of ubiquitinated proteins for autophagosome entry. Casein kinase 2 (CK2) phosphorylates S403 of p62 directly. Furthermore, CK2 overexpression or phosphatase inhibition reduces the formation of inclusion bodies of the polyglutamine-expanded huntingtin exon1 fragment in a p62-dependent manner. We propose that phosphorylation of p62 at S403 regulates autophagic clearance of ubiquitinated proteins and protein aggregates that are poorly degraded by proteasomes.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Autophagy , Serine/genetics , Ubiquitinated Proteins/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Casein Kinase II/genetics , Casein Kinase II/metabolism , Humans , Huntingtin Protein , Mutation , Nerve Tissue Proteins/metabolism , Nuclear Proteins/metabolism , Phosphorylation , Proteasome Endopeptidase Complex/metabolism , Sequestosome-1 Protein , Serine/metabolism , Transfection
7.
J Biol Chem ; 292(32): 13428-13440, 2017 08 11.
Article in English | MEDLINE | ID: mdl-28655765

ABSTRACT

Voltage-gated sodium channels (VGSCs) are transmembrane proteins required for the generation of action potentials in excitable cells and essential for propagating electrical impulses along nerve cells. VGSCs are complexes of a pore-forming α subunit and auxiliary ß subunits, designated as ß1/ß1B-ß4 (encoded by SCN1B-4B, respectively), which also function in cell-cell adhesion. We previously reported the structural basis for the trans homophilic interaction of the ß4 subunit, which contributes to its adhesive function. Here, using crystallographic and biochemical analyses, we show that the ß4 extracellular domains directly interact with each other in a parallel manner that involves an intermolecular disulfide bond between the unpaired Cys residues (Cys58) in the loop connecting strands B and C and intermolecular hydrophobic and hydrogen-bonding interactions of the N-terminal segments (Ser30-Val35). Under reducing conditions, an N-terminally deleted ß4 mutant exhibited decreased cell adhesion compared with the wild type, indicating that the ß4 cis dimer contributes to the trans homophilic interaction of ß4 in cell-cell adhesion. Furthermore, this mutant exhibited increased association with the α subunit, indicating that the cis dimerization of ß4 affects α-ß4 complex formation. These observations provide the structural basis for the parallel dimer formation of ß4 in VGSCs and reveal its mechanism in cell-cell adhesion.


Subject(s)
Models, Molecular , Voltage-Gated Sodium Channel beta-4 Subunit/metabolism , Animals , CHO Cells , Cell Adhesion , Cricetulus , Crystallography, X-Ray , Cysteine/chemistry , Cystine/chemistry , Dimerization , Humans , Hydrogen Bonding , Hydrophobic and Hydrophilic Interactions , Mice , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/metabolism , Protein Conformation , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Protein Multimerization , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Voltage-Gated Sodium Channel beta-4 Subunit/chemistry , Voltage-Gated Sodium Channel beta-4 Subunit/genetics
8.
PLoS Genet ; 11(9): e1005503, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26355680

ABSTRACT

Leucine-rich repeat kinase 2 (LRRK2) is a key molecule in the pathogenesis of familial and idiopathic Parkinson's disease (PD). We have identified two novel LRRK2-associated proteins, a HECT-type ubiquitin ligase, HERC2, and an adaptor-like protein with six repeated Neuralized domains, NEURL4. LRRK2 binds to NEURL4 and HERC2 via the LRRK2 Ras of complex proteins (ROC) domain and NEURL4, respectively. HERC2 and NEURL4 link LRRK2 to the cellular vesicle transport pathway and Notch signaling, through which the LRRK2 complex promotes the recycling of the Notch ligand Delta-like 1 (Dll1)/Delta (Dl) through the modulation of endosomal trafficking. This process negatively regulates Notch signaling through cis-inhibition by stabilizing Dll1/Dl, which accelerates neural stem cell differentiation and modulates the function and survival of differentiated dopaminergic neurons. These effects are strengthened by the R1441G ROC domain-mutant of LRRK2. These findings suggest that the alteration of Notch signaling in mature neurons is a component of PD etiology linked to LRRK2.


Subject(s)
Endosomes/metabolism , Parkinson Disease/enzymology , Protein Serine-Threonine Kinases/physiology , Receptors, Notch/metabolism , Signal Transduction/physiology , Animals , Carrier Proteins/metabolism , Dopamine/metabolism , Drosophila , Guanine Nucleotide Exchange Factors/metabolism , HEK293 Cells , Humans , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2 , Protein Binding , Protein Serine-Threonine Kinases/metabolism , Ubiquitin-Protein Ligases
9.
Hum Mol Genet ; 24(15): 4429-42, 2015 Aug 01.
Article in English | MEDLINE | ID: mdl-25972374

ABSTRACT

Selective autophagy adaptor proteins, including p62/SQSTM1, play pivotal roles in the targeted degradation of ubiquitinated proteins or organelles through the autophagy-lysosome system. However, how autophagy adaptors promote the autophagosomal engulfment of selected substrates is poorly understood. Here, we show that p62 phosphorylation at S403 is required for the efficient autophagosomal engulfment of polyubiquitinated mitochondria during Parkin-dependent mitophagy. p62 is able to interact with Parkin-recruited mitochondria without S403 phosphorylation under mitophagy-inducing conditions, but those mitochondria are not enclosed by autophagosomes. Intriguingly, the S403 phosphorylation occurs only in the early period of mitochondrial depolarization. Optineurin and TANK-binding kinase 1 (TBK1) are transiently recruited to the polyubiquitinated mitochondria, and the activated TBK1 phosphorylates p62 at S403. TBK1 inhibitor, BX795, prevents the p62-mediated autophagosomal engulfment of Parkin-recruited mitochondria. Our results suggest that TBK1-mediated S403 phosphorylation regulates the efficient autophagosomal engulfment of ubiquitinated mitochondria as an immediate response to the mitochondrial depolarization.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Autophagy/genetics , Mitochondria/genetics , Protein Serine-Threonine Kinases/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cell Line , Humans , Mice , Mice, Knockout , Mitochondria/metabolism , Mitophagy/genetics , Phosphorylation , Polyubiquitin/metabolism , Protein Serine-Threonine Kinases/metabolism , Sequestosome-1 Protein , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism
10.
Hum Mol Genet ; 24(3): 740-56, 2015 Feb 01.
Article in English | MEDLINE | ID: mdl-25274774

ABSTRACT

In some neurological diseases caused by repeat expansions such as myotonic dystrophy, the RNA-binding protein muscleblind-like 1 (MBNL1) accumulates in intranuclear inclusions containing mutant repeat RNA. The interaction between MBNL1 and mutant RNA in the nucleus is a key event leading to loss of MBNL function, yet the details of this effect have been elusive. Here, we investigated the mechanism and significance of MBNL1 nuclear localization. We found that MBNL1 contains two classes of nuclear localization signal (NLS), a classical bipartite NLS and a novel conformational NLS. Alternative splicing of exon 7 acts as a switch between these NLS types and couples MBNL1 activity and intracellular localization. Depending on its nuclear localization, MBNL1 promoted nuclear accumulation of mutant RNA containing a CUG or CAG repeat, some of which produced proteins containing homopolymeric tracts such as polyglutamine. Furthermore, MBNL1 repressed the expression of these homopolymeric proteins including those presumably produced through repeat-associated non-ATG (RAN) translation. These results suggest that nuclear retention of expanded RNA reflects a novel role of MBNL proteins in repressing aberrant protein expression and may provide pathological and therapeutic implications for a wide range of repeat expansion diseases associated with nuclear RNA retention and/or RAN translation.


Subject(s)
Cell Nucleus/metabolism , DNA-Binding Proteins/metabolism , Nuclear Localization Signals/genetics , RNA, Messenger/metabolism , RNA-Binding Proteins/metabolism , Trinucleotide Repeat Expansion , Alternative Splicing , Animals , COS Cells , Cell Line, Tumor , Cell Nucleus/genetics , Chlorocebus aethiops , DNA-Binding Proteins/chemistry , Gene Expression Regulation , Humans , Mice , Mutation , Nuclear Localization Signals/metabolism , RNA, Messenger/genetics , RNA-Binding Proteins/chemistry
11.
Hum Mol Genet ; 24(4): 1092-105, 2015 Feb 15.
Article in English | MEDLINE | ID: mdl-25305080

ABSTRACT

Huntington's disease (HD) is a dominantly inherited genetic disease caused by mutant huntingtin (htt) protein with expanded polyglutamine (polyQ) tracts. A neuropathological hallmark of HD is the presence of neuronal inclusions of mutant htt. p62 is an important regulatory protein in selective autophagy, a process by which aggregated proteins are degraded, and it is associated with several neurodegenerative disorders including HD. Here, we investigated the effect of p62 depletion in three HD model mice: R6/2, HD190QG and HD120QG mice. We found that loss of p62 in these models led to longer life spans and reduced nuclear inclusions, although cytoplasmic inclusions increased with polyQ length. In mouse embryonic fibroblasts (MEFs) with or without p62, mutant htt with a nuclear localization signal (NLS) showed no difference in nuclear inclusion between the two MEF types. In the case of mutant htt without NLS, however, p62 depletion increased cytoplasmic inclusions. Furthermore, to examine the effect of impaired autophagy in HD model mice, we crossed R6/2 mice with Atg5 conditional knockout mice. These mice also showed decreased nuclear inclusions and increased cytoplasmic inclusions, similar to HD mice lacking p62. These data suggest that the genetic ablation of p62 in HD model mice enhances cytoplasmic inclusion formation by interrupting autophagic clearance of polyQ inclusions. This reduces polyQ nuclear influx and paradoxically ameliorates disease phenotypes by decreasing toxic nuclear inclusions.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Huntington Disease/genetics , Intranuclear Inclusion Bodies/genetics , Phenotype , Animals , Autophagy , Corpus Striatum/metabolism , Corpus Striatum/pathology , Disease Models, Animal , Female , Hippocampus/metabolism , Hippocampus/pathology , Huntington Disease/mortality , Huntington Disease/pathology , Intracellular Space/metabolism , Longevity/genetics , Mice , Mice, Knockout , Peptides/genetics , Proteolysis
12.
Proc Natl Acad Sci U S A ; 111(38): E3966-75, 2014 Sep 23.
Article in English | MEDLINE | ID: mdl-25201980

ABSTRACT

The inositol 1,4,5-trisphosphate receptor (IP3R) in the endoplasmic reticulum mediates calcium signaling that impinges on intracellular processes. IP3Rs are allosteric proteins comprising four subunits that form an ion channel activated by binding of IP3 at a distance. Defective allostery in IP3R is considered crucial to cellular dysfunction, but the specific mechanism remains unknown. Here we demonstrate that a pleiotropic enzyme transglutaminase type 2 targets the allosteric coupling domain of IP3R type 1 (IP3R1) and negatively regulates IP3R1-mediated calcium signaling and autophagy by locking the subunit configurations. The control point of this regulation is the covalent posttranslational modification of the Gln2746 residue that transglutaminase type 2 tethers to the adjacent subunit. Modification of Gln2746 and IP3R1 function was observed in Huntington disease models, suggesting a pathological role of this modification in the neurodegenerative disease. Our study reveals that cellular signaling is regulated by a new mode of posttranslational modification that chronically and enzymatically blocks allosteric changes in the ligand-gated channels that relate to disease states.


Subject(s)
Autophagy , Calcium Signaling , GTP-Binding Proteins/metabolism , Huntington Disease/metabolism , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Protein Processing, Post-Translational , Transglutaminases/metabolism , Allosteric Regulation/genetics , Animals , Disease Models, Animal , GTP-Binding Proteins/genetics , HeLa Cells , Humans , Huntington Disease/genetics , Huntington Disease/pathology , Inositol 1,4,5-Trisphosphate Receptors/genetics , PC12 Cells , Protein Glutamine gamma Glutamyltransferase 2 , Protein Structure, Tertiary , Rats , Transglutaminases/genetics
13.
PLoS Genet ; 10(12): e1004861, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25474007

ABSTRACT

The kinase PINK1 and the E3 ubiquitin (Ub) ligase Parkin participate in mitochondrial quality control. The phosphorylation of Ser65 in Parkin's ubiquitin-like (UBl) domain by PINK1 stimulates Parkin activation and translocation to damaged mitochondria, which induces mitophagy generating polyUb chain. However, Parkin Ser65 phosphorylation is insufficient for Parkin mitochondrial translocation. Here we report that Ser65 in polyUb chain is also phosphorylated by PINK1, and that phosphorylated polyUb chain on mitochondria tethers Parkin at mitochondria. The expression of Tom70MTS-4xUb SE, which mimics phospho-Ser65 polyUb chains on the mitochondria, activated Parkin E3 activity and its mitochondrial translocation. An E3-dead form of Parkin translocated to mitochondria with reduced membrane potential in the presence of Tom70(MTS)-4xUb SE, whereas non-phospho-polyUb mutant Tom70(MTS)-4xUb SA abrogated Parkin translocation. Parkin binds to the phospho-polyUb chain through its RING1-In-Between-RING (IBR) domains, but its RING0-linker is also required for mitochondrial translocation. Moreover, the expression of Tom70(MTS)-4xUb SE improved mitochondrial degeneration in PINK1-deficient, but not Parkin-deficient, Drosophila. Our study suggests that the phosphorylation of mitochondrial polyUb by PINK1 is implicated in both Parkin activation and mitochondrial translocation, predicting a chain reaction mechanism of mitochondrial phospho-polyUb production by which rapid translocation of Parkin is achieved.


Subject(s)
Mitochondria/metabolism , Polyubiquitin/metabolism , Protein Kinases/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , Animals, Genetically Modified , Cells, Cultured , Drosophila , HEK293 Cells , HeLa Cells , Humans , Mice , Phosphorylation , Protein Binding , Protein Transport , Ubiquitination
14.
Genesis ; 54(11): 568-572, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27596971

ABSTRACT

VAChT-Cre.Fast and VAChT-Cre.Slow mice selectively express Cre recombinase in approximately one half of postnatal somatic motor neurons. The mouse lines have been used in various studies with selective genetic modifications in adult motor neurons. In the present study, we crossed VAChT-Cre lines with a reporter line, CAG-Syp/tdTomato, in which synaptophysin-tdTomato fusion proteins are efficiently sorted to axon terminals, making it possible to label both cell bodies and axon terminals of motor neurons. In the mice, Syp/tdTomato fluorescence preferentially co-localized with osteopontin, a recently discovered motor neuron marker for slow-twitch fatigue-resistant (S) and fast-twitch fatigue-resistant (FR) types. The fluorescence did not preferentially co-localize with matrix metalloproteinase-9, a marker for fast-twitch fatigable (FF) motor neurons. In the neuromuscular junctions, Syp/tdTomato fluorescence was detected mainly in motor nerve terminals that innervate type I or IIa muscle fibers. These results suggest that the VAChT-Cre lines are Cre-drivers that have selectivity in S and FR motor neurons. In order to avoid confusion, we have changed the mouse line names from VAChT-Cre.Fast and VAChT-Cre.Slow to VAChT-Cre.Early and VAChT-Cre.Late, respectively. The mouse lines will be useful tools to study slow-type motor neurons, in relation to physiology and pathology.


Subject(s)
Axons/metabolism , Motor Neurons/metabolism , Muscle Fibers, Fast-Twitch/metabolism , Vesicular Acetylcholine Transport Proteins/genetics , Animals , Integrases/genetics , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinase 9/metabolism , Mice , Osteopontin/genetics , Synaptophysin/genetics , Synaptophysin/metabolism , Vesicular Acetylcholine Transport Proteins/metabolism
15.
Hum Mol Genet ; 23(10): 2737-51, 2014 May 15.
Article in English | MEDLINE | ID: mdl-24381308

ABSTRACT

Huntington's disease (HD) is an inherited neurodegenerative disorder caused by abnormal expansion of glutamine repeats in the protein huntingtin. In HD brain, mutant huntingtin undergoes proteolytic processing, and its N-terminal fragment containing poly-glutamine repeats accumulate as insoluble aggregates leading to the defect in cellular protein quality control system and heat shock response (HSR). Here we demonstrate that the defective HSR in the brain is due to the down-regulation of heat shock factor 1 (HSF1) in both mice and fly models of HD. Interestingly, treatment of dexamethasone (a synthetic glucocorticoid) to HD mice or flies significantly increased the expression and transactivation of HSF1 and induction of HSR and these effects are mediated through the down-regulation of HSP90. Dexamethasone treatment also significantly decreased the aggregate load and transient recovery of HD-related behavioural phenotypes in both disease models. These results suggest that dexamethasone could be a potential therapeutic molecule for the treatment of HD and related poly-glutamine disorders.


Subject(s)
Dexamethasone/pharmacology , Glucocorticoids/pharmacology , Heat-Shock Response/drug effects , Huntington Disease/drug therapy , Animals , Brain/metabolism , Cell Line , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Dexamethasone/therapeutic use , Disease Models, Animal , Disease Progression , Drosophila , Drug Evaluation, Preclinical , Female , Glucocorticoids/therapeutic use , HSP70 Heat-Shock Proteins/metabolism , Heat Shock Transcription Factors , Humans , Huntington Disease/metabolism , Male , Mice, Inbred C57BL , Mice, Transgenic , Psychomotor Performance/drug effects , Transcription Factors/genetics , Transcription Factors/metabolism , Transcriptional Activation
16.
J Biol Chem ; 289(2): 1192-202, 2014 Jan 10.
Article in English | MEDLINE | ID: mdl-24280224

ABSTRACT

Dominant mutations in FUS/TLS cause a familial form of amyotrophic lateral sclerosis (fALS), where abnormal accumulation of mutant FUS proteins in cytoplasm has been observed as a major pathological change. Many of pathogenic mutations have been shown to deteriorate the nuclear localization signal in FUS and thereby facilitate cytoplasmic mislocalization of mutant proteins. Several other mutations, however, exhibit no effects on the nuclear localization of FUS in cultured cells, and their roles in the pathomechanism of fALS remain obscure. Here, we show that a pathogenic mutation, G156E, significantly increases the propensities for aggregation of FUS in vitro and in vivo. Spontaneous in vitro formation of amyloid-like fibrillar aggregates was observed in mutant but not wild-type FUS, and notably, those fibrils functioned as efficient seeds to trigger the aggregation of wild-type protein. In addition, the G156E mutation did not disturb the nuclear localization of FUS but facilitated the formation of intranuclear inclusions in rat hippocampal neurons with significant cytotoxicity. We thus propose that intranuclear aggregation of FUS triggered by a subset of pathogenic mutations is an alternative pathomechanism of FUS-related fALS diseases.


Subject(s)
Amyotrophic Lateral Sclerosis/metabolism , Cell Nucleus/metabolism , Mutation , RNA-Binding Protein FUS/metabolism , Amino Acid Sequence , Amino Acid Substitution , Amyloid/chemistry , Amyloid/metabolism , Amyloid/ultrastructure , Amyotrophic Lateral Sclerosis/genetics , Animals , Cell Line, Tumor , Cells, Cultured , Electrophoresis, Polyacrylamide Gel , Glutathione Transferase/genetics , Glutathione Transferase/metabolism , Humans , Microscopy, Confocal , Microscopy, Electron , Neurons/cytology , Neurons/metabolism , Protein Denaturation , RNA-Binding Protein FUS/chemistry , RNA-Binding Protein FUS/genetics , Rats , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism
17.
Biochim Biophys Acta ; 1842(9): 1472-84, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24769000

ABSTRACT

Polyglutamine diseases are a family of inherited neurodegenerative diseases caused by the expansion of CAG repeats within the coding region of target genes. Still the mechanism(s) by which polyglutamine proteins are ubiquitinated and degraded remains obscure. Here, for the first time, we demonstrate that Mahogunin 21 ring finger 1 E3 ubiquitin protein ligase is depleted in cells that express expanded-polyglutamine proteins. MGRN1 co-immunoprecipitates with expanded-polyglutamine huntingtin and ataxin-3 proteins. Furthermore, we show that MGRN1 is predominantly colocalized and recruits with polyglutamine aggregates in both cellular and transgenic mouse models. Finally, we demonstrate that the partial depletion of MGRN1 increases the rate of aggregate formation and cell death, whereas the overexpression of MGRN1 reduces the frequency of aggregate formation and provides cytoprotection against polyglutamine-induced proteotoxicity. These observations suggest that stimulating the activity of MGRN1 ubiquitin ligase might be a potential therapeutic target to eliminate the cytotoxic threat in polyglutamine diseases.


Subject(s)
Apoptosis , Nerve Tissue Proteins/metabolism , Peptides/metabolism , Protein Folding , Ubiquitin-Protein Ligases/physiology , Ubiquitin/metabolism , Animals , Blotting, Western , Cell Proliferation , Cells, Cultured , Fluorescent Antibody Technique , Humans , Huntingtin Protein , Immunoprecipitation , Male , Mice , Mice, Transgenic , Molecular Chaperones/genetics , Molecular Chaperones/metabolism , Nerve Tissue Proteins/genetics , Proteasome Endopeptidase Complex/metabolism , Proteolysis , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction
18.
Biochem Biophys Res Commun ; 467(2): 322-7, 2015 Nov 13.
Article in English | MEDLINE | ID: mdl-26454170

ABSTRACT

The tumor suppressor p53, a 393-amino acid transcription factor with four domains, induces cell cycle arrest, senescence, and apoptosis in response to diverse stress. Tetramer formation is critical for the function of p53. The tetramerization domain permits the tetramerization of p53, where bundled four DNA-binding domains recognize the specific target DNA sequences and activate hundreds of genes, which lead to the various cell fates. Here we show that tumor suppressive functions of p53 can be regulated by manipulating tetramer formation of an engineered p53, in which tetramerization domain of p53 is replaced with an inducible tetramer forming protein. This result suggests that artificial regulation of p53 activity by the engineered p53 is a useful tool to investigate the tumor suppression mechanism of p53 and to combat cancer.


Subject(s)
Recombinant Fusion Proteins/chemistry , TOR Serine-Threonine Kinases/chemistry , Tacrolimus Binding Proteins/chemistry , Tumor Suppressor Protein p53/chemistry , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , Humans , Plasmids/chemistry , Plasmids/metabolism , Protein Engineering , Protein Interaction Domains and Motifs , Protein Multimerization , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism , Tacrolimus Binding Proteins/genetics , Tacrolimus Binding Proteins/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
19.
Biochem Biophys Res Commun ; 463(4): 1196-202, 2015 Aug 07.
Article in English | MEDLINE | ID: mdl-26086102

ABSTRACT

Dominant mutations in Cu,Zn-superoxide dismutase (SOD1) cause a familial form of amyotrophic lateral sclerosis (ALS). A pathological hallmark of the familial ALS is the formation of mutant SOD1 aggregates, leading to the proposal that SOD1 gains toxicities through protein misfolding triggered by mutations. Nevertheless, molecular requirements for mutant SOD1 to acquire pathogenicity still remain obscure. Here, we show that Cys residues in SOD1 are essential to exerting toxicities of SOD1 in a Caenorhabditis elegans model. Exogenous expression of wild-type as well as pathogenic mutant SOD1 fused with a fluorescent protein in C. elegans resulted in the accumulation of disulfide-reduced SOD1 and retarded the worm's motility. In contrast, little effects of exogenously expressed SOD1 on the motility were observed when all four Cys residues in SOD1 were replaced with Ser. Taken together, we propose that deregulation of Cys chemistry in SOD1 proteins is involved in the pathogenesis of SOD1-related ALS.


Subject(s)
Amyotrophic Lateral Sclerosis/enzymology , Caenorhabditis elegans/drug effects , Cysteine/metabolism , Superoxide Dismutase/metabolism , Animals , Disease Models, Animal , Superoxide Dismutase/chemistry
20.
Proc Natl Acad Sci U S A ; 109(45): 18577-82, 2012 Nov 06.
Article in English | MEDLINE | ID: mdl-23090990

ABSTRACT

Voltage-gated sodium (Na(V)) and potassium (K(V)) channels are critical components of neuronal action potential generation and propagation. Here, we report that Na(V)ß1 encoded by SCN1b, an integral subunit of Na(V) channels, coassembles with and modulates the biophysical properties of K(V)1 and K(V)7 channels, but not K(V)3 channels, in an isoform-specific manner. Distinct domains of Na(V)ß1 are involved in modulation of the different K(V) channels. Studies with channel chimeras demonstrate that Na(V)ß1-mediated changes in activation kinetics and voltage dependence of activation require interaction of Na(V)ß1 with the channel's voltage-sensing domain, whereas changes in inactivation and deactivation require interaction with the channel's pore domain. A molecular model based on docking studies shows Na(V)ß1 lying in the crevice between the voltage-sensing and pore domains of K(V) channels, making significant contacts with the S1 and S5 segments. Cross-modulation of Na(V) and K(V) channels by Na(V)ß1 may promote diversity and flexibility in the overall control of cellular excitability and signaling.


Subject(s)
Potassium Channels, Voltage-Gated/metabolism , Voltage-Gated Sodium Channel beta-1 Subunit/metabolism , Animals , CHO Cells , Cricetinae , Cricetulus , Ion Channel Gating , Kinetics , Mice , Models, Molecular , PC12 Cells , Potassium Channels, Voltage-Gated/chemistry , Protein Binding , Protein Structure, Tertiary , Rats , Xenopus
SELECTION OF CITATIONS
SEARCH DETAIL