Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 47
1.
J Biol Chem ; 300(2): 105629, 2024 Feb.
Article En | MEDLINE | ID: mdl-38199563

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.


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.
Biol Reprod ; 110(3): 465-475, 2024 Mar 13.
Article En | MEDLINE | ID: mdl-37995271

The mammalian oviductal lumen is a specialized chamber that provides an environment that strictly regulates fertilization and early embryogenesis, but the regulatory mechanisms to gametes and zygotes are unclear. We evaluated the oviductal regulation of early embryonic development using Ovgp1 (encoding an oviductal humoral factor, OVGP1)-knockout golden hamsters. The experimental results revealed the following: (1) female Ovgp1-knockout hamsters failed to produce litters; (2) in the oviducts of Ovgp1-knockout animals, fertilized eggs were sometimes identified, but their morphology showed abnormal features; (3) the number of implantations in the Ovgp1-knockout females was low; (4) even if implantations occurred, the embryos developed abnormally and eventually died; and (5) Ovgp1-knockout female ovaries transferred to wild-type females resulted in the production of Ovgp1-knockout egg-derived OVGP1-null litters, but the reverse experiment did not. These results suggest that OVGP1-mediated physiological events are crucial for reproductive process in vivo, from fertilization to early embryonic development. This animal model shows that the fate of the zygote is determined not only genetically, but also by the surrounding oviductal microenvironment.


Fallopian Tubes , Oviducts , Humans , Pregnancy , Animals , Cricetinae , Female , Mesocricetus , Germ Cells , Ovary , Mammals , Glycoproteins
3.
Int J Mol Sci ; 22(14)2021 Jul 12.
Article En | MEDLINE | ID: mdl-34299063

Regulatory T cells (Tregs) suppress immune responses and maintain immunological self-tolerance and homeostasis. We currently investigated relationships between skin barrier condition and Treg behavior using skin barrier-disrupted mice. Skin barrier disruption was induced by repeated topical application of 4% sodium dodecyl sulfate (SDS) on mice. The number of CD4+ forkhead box protein P3 (Foxp3)+ Tregs was higher in 4% SDS-treated skins than in controls. This increasing was correlated with the degree of acanthosis. The numbers of interleukin (IL)-10+ and transforming growth factor (TGF)-ß+ Tregs also increased in 4% SDS-treated skins. Localization of IL-33 in keratinocytes shifted from nucleus to cytoplasm after skin barrier disruption. Notably, IL-33 promoted the migration of Tregs in chemotaxis assay. The skin infiltration of Tregs was cancelled in IL-33 neutralizing antibody-treated mice and IL-33 knockout mice. Thus, keratinocyte-derived IL-33 may induce Treg migration into barrier-disrupted skin to control the phase transition between healthy and inflammatory conditions.


Cell Movement , Chemotaxis , Dermatitis/pathology , Interleukin-33/physiology , Skin/pathology , T-Lymphocytes, Regulatory/immunology , Animals , Dermatitis/immunology , Dermatitis/metabolism , Forkhead Transcription Factors/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Skin/metabolism
4.
J Biol Chem ; 295(29): 9768-9785, 2020 07 17.
Article En | MEDLINE | ID: mdl-32499373

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.


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
5.
J Histochem Cytochem ; 67(11): 813-824, 2019 11.
Article En | MEDLINE | ID: mdl-31424977

The arrangement of immature germ cells changes regularly and periodically along the axis of the seminiferous tubule, and is used to describe the progression of spermatogenesis. This description is based primarily on the changes in the acrosome and the nuclear morphology of haploid spermatids. However, such criteria cannot be applied under pathological conditions with arrested spermatid differentiation. In such settings, the changes associated with the differentiation of premeiotic germ cells must be analyzed. Here, we found that the unique bipolar motor protein, KIF11 (kinesin-5/Eg5), which functions in spindle formation during mitosis and meiosis in oocytes and early embryos, is expressed in premeiotic germ cells (spermatogonia and spermatocytes). Thus, we aimed to investigate whether KIF11 could be used to describe the progression of incomplete spermatogenesis. Interestingly, KIF11 expression was barely observed in haploid spermatids and Sertoli cells. The KIF11 staining allowed us to evaluate the progression of meiotic processes, by providing the time axis of spindle formation in both normal and spermatogenesis-arrested mutant mice. Accordingly, KIF11 has the potential to serve as an excellent marker to describe spermatogenesis, even in the absence of spermatid development.


Kinesins/analysis , Seminiferous Tubules/cytology , Spermatogenesis , Animals , Male , Meiosis , Mice , Mice, Inbred C57BL , Seminiferous Tubules/ultrastructure , Spermatids/cytology , Spermatocytes/cytology , Spermatogonia/cytology
6.
Acta Neuropathol Commun ; 6(1): 96, 2018 09 19.
Article En | MEDLINE | ID: mdl-30231908

Accumulating evidence suggests that the lesions of Parkinson's disease (PD) expand due to transneuronal spreading of fibrils composed of misfolded alpha-synuclein (a-syn), over the course of 5-10 years. However, the precise mechanisms and the processes underlying the spread of these fibril seeds have not been clarified in vivo. Here, we investigated the speed of a-syn transmission, which has not been a focus of previous a-syn transmission experiments, and whether a-syn pathologies spread in a neural circuit-dependent manner in the mouse brain. We injected a-syn preformed fibrils (PFFs), which are seeds for the propagation of a-syn deposits, either before or after callosotomy, to disconnect bilateral hemispheric connections. In mice that underwent callosotomy before the injection, the propagation of a-syn pathology to the contralateral hemisphere was clearly reduced. In contrast, mice that underwent callosotomy 24 h after a-syn PFFs injection showed a-syn pathology similar to that seen in mice without callosotomy. These results suggest that a-syn seeds are rapidly disseminated through neuronal circuits immediately after seed injection, in a prion-like seeding experiment in vivo, although it is believed that clinical a-syn pathologies take years to spread throughout the brain. In addition, we found that botulinum toxin B blocked the transsynaptic transmission of a-syn seeds by specifically inactivating the synaptic vesicle fusion machinery. This study offers a novel concept regarding a-syn propagation, based on the Braak hypothesis, and also cautions that experimental transmission systems may be examining a unique type of transmission, which differs from the clinical disease state.


Brain/pathology , Nerve Net/pathology , Neurons/metabolism , Parkinson Disease/pathology , Prion Diseases/pathology , alpha-Synuclein/metabolism , Amyloid/toxicity , Animals , Botulinum Toxins, Type A/metabolism , Dopamine and cAMP-Regulated Phosphoprotein 32/metabolism , Functional Laterality , Humans , Mice, Inbred C57BL , NAV1.2 Voltage-Gated Sodium Channel/metabolism , Neurons/pathology , Protein Transport , Synaptic Vesicles/drug effects , Synaptic Vesicles/physiology , Time Factors , Tyrosine 3-Monooxygenase/metabolism
7.
PLoS One ; 13(8): e0200988, 2018.
Article En | MEDLINE | ID: mdl-30086173

Although the aggregation of amyloid-ß peptide (Aß) clearly plays a central role in the pathogenesis of Alzheimer's disease (AD), endosomal traffic dysfunction is considered to precede Aß aggregation and trigger AD pathogenesis. A body of evidence suggests that the ß-carboxyl-terminal fragment (ßCTF) of amyloid-ß precursor protein (APP), which is the direct precursor of Aß, accumulates in endosomes and causes vesicular traffic impairment. However, the mechanism underlying this impairment remains unclear. Here we identified TMEM30A as a candidate partner for ßCTF. TMEM30A is a subcomponent of lipid flippase that translocates phospholipids from the outer to the inner leaflet of the lipid bilayer. TMEM30A physically interacts with ßCTF in endosomes and may impair vesicular traffic, leading to abnormally enlarged endosomes. APP traffic is also concomitantly impaired, resulting in the accumulation of APP-CTFs, including ßCTF. In addition, we found that expressed BACE1 accumulated in enlarged endosomes and increased Aß production. Our data suggested that TMEM30A is involved in ßCTF-dependent endosome abnormalities that are related to Aß overproduction.


Alzheimer Disease/metabolism , Amyloid beta-Protein Precursor/chemistry , Amyloid beta-Protein Precursor/metabolism , Endosomes/metabolism , Membrane Proteins/chemistry , Membrane Proteins/metabolism , Alzheimer Disease/genetics , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Protein Precursor/genetics , Animals , Aspartic Acid Endopeptidases/metabolism , COS Cells , Chlorocebus aethiops , Endosomes/pathology , Humans , Membrane Proteins/genetics , Multiprotein Complexes/chemistry , Multiprotein Complexes/genetics , Multiprotein Complexes/metabolism , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/metabolism , Protein Interaction Domains and Motifs , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
8.
J Clin Invest ; 127(4): 1438-1450, 2017 Apr 03.
Article En | MEDLINE | ID: mdl-28263187

Huntington's disease (HD) is a polyglutamine (polyQ) disease caused by aberrant expansion of the polyQ tract in Huntingtin (HTT). While motor impairment mediated by polyQ-expanded HTT has been intensively studied, molecular mechanisms for nonmotor symptoms in HD, such as psychiatric manifestations, remain elusive. Here we have demonstrated that HTT forms a ternary protein complex with the scaffolding protein DISC1 and cAMP-degrading phosphodiesterase 4 (PDE4) to regulate PDE4 activity. We observed pathological cross-seeding between DISC1 and mutant HTT aggregates in the brains of HD patients as well as in a murine model that recapitulates the polyQ pathology of HD (R6/2 mice). In R6/2 mice, consequent reductions in soluble DISC1 led to dysregulation of DISC1-PDE4 complexes, aberrantly increasing the activity of PDE4. Importantly, exogenous expression of a modified DISC1, which binds to PDE4 but not mutant HTT, normalized PDE4 activity and ameliorated anhedonia in the R6/2 mice. We propose that cross-seeding of mutant HTT and DISC1 and the resultant changes in PDE4 activity may underlie the pathology of a specific subset of mental manifestations of HD, which may provide an insight into molecular signaling in mental illness in general.


Cyclic Nucleotide Phosphodiesterases, Type 4/metabolism , Huntington Disease/enzymology , Nerve Tissue Proteins/metabolism , Protein Aggregation, Pathological/enzymology , Animals , Female , HEK293 Cells , Humans , Huntingtin Protein/genetics , Huntingtin Protein/metabolism , Mice, Transgenic , Mutation
9.
Sci Rep ; 6: 35236, 2016 10 14.
Article En | MEDLINE | ID: mdl-27739513

FUS/TLS is an RNA/DNA-binding protein associated with neurodegenerative diseases including amyotrophic lateral sclerosis and frontotemporal lobar degeneration. Previously, we found that a prion-like domain in the N-terminus of FUS/TLS mediates co-aggregation between FUS/TLS and mutant huntingtin, the gene product of Huntington's disease (HD). Here, we show that heterozygous knockout of FUS/TLS worsened the phenotypes of model mice of (HD, but not spinal and bulbar muscular atrophy (SBMA). This difference was correlated with the degree of pathological association between disease proteins and FUS/TLS. Co-aggregation between FUS/TLS and mutant huntingtin resulted in the depletion of free FUS/TLS protein in HD mice that was detected as a monomer in SDS-PAGE analysis. Recently, we found that FUS/TLS paralogs, TAF15 and EWS, were up-regulated in homozygous FUS/TLS knockout mice. These two proteins were up-regulated in both HD and FUS/TLS heterozygote mice, and were further elevated in HD-TLS+/- double mutant mice, consistent with the functional impairment of FUS/TLS. These results suggest that FUS/TLS sequestration by co-aggregation is a rate-limiting factor of disease phenotypes of HD and that inclusions may have an adverse aspect, rather than being simply benign or protective. In addition, our results highlight inclusions as repositories of potential modifiers of neurodegeneration.


Neurodegenerative Diseases/metabolism , Peptides/metabolism , RNA-Binding Protein FUS/metabolism , Animals , DNA-Binding Proteins/metabolism , Disease Models, Animal , Female , Homozygote , Huntingtin Protein/metabolism , Inclusion Bodies/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic/metabolism , Mutation/physiology , Phenotype , RNA-Binding Proteins/metabolism , Up-Regulation/physiology
10.
Sci Rep ; 6: 34575, 2016 Sep 30.
Article En | MEDLINE | ID: mdl-27687130

The mammalian central nervous system (CNS) contains various types of neurons with different neuronal functions. In contrast to established roles of cell type-specific transcription factors on neuronal specification and maintenance, whether ubiquitous transcription factors have conserved or differential neuronal function remains uncertain. Here, we revealed that inactivation of a ubiquitous factor NF-Y in different sets of neurons resulted in cell type-specific neuropathologies and gene downregulation in mouse CNS. In striatal and cerebellar neurons, NF-Y inactivation led to ubiquitin/p62 pathologies with downregulation of an endoplasmic reticulum (ER) chaperone Grp94, as we previously observed by NF-Y deletion in cortical neurons. In contrast, NF-Y inactivation in motor neurons induced neuronal loss without obvious protein deposition. Detailed analysis clarified downregulation of another ER chaperone Grp78 in addition to Grp94 in motor neurons, and knockdown of both ER chaperones in motor neurons recapitulated the pathology observed after NF-Y inactivation. Finally, additional downregulation of Grp78 in striatal neurons suppressed ubiquitin accumulation induced by NF-Y inactivation, implying that selective ER chaperone downregulation mediates different neuropathologies. Our data suggest distinct roles of NF-Y in protein homeostasis and neuronal maintenance in the CNS by differential regulation of ER chaperone expression.

11.
FEBS J ; 283(6): 1077-87, 2016 Mar.
Article En | MEDLINE | ID: mdl-26756308

UNLABELLED: The upstream transcription factors (USFs) USF1 and USF2 are ubiquitously expressed transcription factors that are characterized by a conserved basic helix-loop-helix/leucine zipper DNA-binding domain. They form homo- or heterodimers, and recognize E-box motifs to modulate gene expression. They are known to regulate diverse cellular functions, including the cell cycle, immune responses and glucose/lipid metabolism, but their roles in neuronal cells remain to be clarified. Here, we performed chromatin immunoprecipitation of USF1 from mouse brain cortex. Subsequent promoter array analysis (ChIP-chip) indicated that USF1 exclusively bound to the CACGTG E-box motifs in the proximal promoter regions. Importantly, functional annotation of the USF1-binding targets revealed an enrichment of genes related to lysosomal functions. Gene expression array analysis using a neuronal cell line subsequently revealed that knockdown of USFs de-regulated lysosomal gene expression. Altered expression was validated by quantitative RT-PCR, supporting the conclusion that USFs regulate lysosomal gene expression. Furthermore, USF knockdown slightly increased LysoTracker Red staining, implying a role for USFs in modulating lysosomal homeostasis. Together, our comprehensive genome-scale analyses identified lysosomal genes as targets of USFs in neuronal cells, suggesting a potential additional pathway of lysosomal regulation. DATABASE: The data for the gene expression array and ChIP-chip have been submitted to the Gene Expression Omnibus (GEO) under accession numbers GSE76615 and GSE76616, respectively.


Lysosomes/genetics , Lysosomes/metabolism , Neurons/metabolism , Upstream Stimulatory Factors/metabolism , Animals , Binding Sites/genetics , Cell Line , Cerebral Cortex/metabolism , Chromatin Immunoprecipitation , E-Box Elements , Gene Expression Regulation , Gene Knockdown Techniques , Genome-Wide Association Study , Male , Mice , Mice, Inbred CBA , Oligonucleotide Array Sequence Analysis , Promoter Regions, Genetic , Upstream Stimulatory Factors/antagonists & inhibitors , Upstream Stimulatory Factors/genetics
12.
Neurosci Res ; 103: 64-70, 2016 Feb.
Article En | MEDLINE | ID: mdl-26302676

Protein inclusions in neurodegenerative diseases are associated with p62, which has an important role in autophagic clearance of polyubiquitinated proteins. Selective autophagy is regulated by S403-phosphorylation of p62, and S403-phosphorylated p62 (S403-phos-p62) accumulates in Atg5 conditional knockout (Atg5CKO) mice in which autophagosome formation is impaired. We performed immunohistochemical tests for the presence of S403-phos-p62 in postmortem brain of neurodegenerative disease cases, and found accumulations in amyotrophic lateral sclerosis and Alzheimer's disease tissues. In Atg5CKO and HD190QG (Huntington's disease model) mice, however, we found a postmortem decrease in S403-phos-p62 immunoreactivity, suggesting that post-mortem changes should be considered when interpreting human data.


Adaptor Proteins, Signal Transducing/metabolism , Alzheimer Disease/metabolism , Amyotrophic Lateral Sclerosis/metabolism , Heat-Shock Proteins/metabolism , Huntington Disease/metabolism , Intranuclear Inclusion Bodies/metabolism , Aged , Aged, 80 and over , Animals , Brain/metabolism , Humans , Huntingtin Protein , Huntington Disease/genetics , Mice, Mutant Strains , Middle Aged , Nerve Tissue Proteins/genetics , Nuclear Proteins/genetics , Phosphorylation , Postmortem Changes , Sequestosome-1 Protein
13.
Biochem Biophys Res Commun ; 463(4): 1196-202, 2015 Aug 07.
Article En | MEDLINE | ID: mdl-26086102

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.


Amyotrophic Lateral Sclerosis/enzymology , Caenorhabditis elegans/drug effects , Cysteine/metabolism , Superoxide Dismutase/metabolism , Animals , Disease Models, Animal , Superoxide Dismutase/chemistry
14.
Acta Neuropathol Commun ; 3: 24, 2015 Apr 25.
Article En | MEDLINE | ID: mdl-25907258

INTRODUCTION: FUS/TLS is an RNA-binding protein whose genetic mutations or pathological inclusions are associated with neurological diseases including amyotrophic lateral sclerosis (ALS), frontotemporal lobar degeneration, and essential tremor (ET). It is unclear whether their pathogenesis is mediated by gain or loss of function of FUS/TLS. RESULTS: Here, we established outbred FUS/TLS knockout mice to clarify the effects of FUS/TLS dysfunction in vivo. We obtained homozygous knockout mice that grew into adulthood. Importantly, they did not manifest ALS- or ET-like phenotypes until nearly two years. Instead, they showed distinct histological and behavioral alterations including vacuolation in hippocampus, hyperactivity, and reduction in anxiety-like behavior. Knockout mice showed transcriptome alterations including upregulation of Taf15 and Hnrnpa1, while they have normal morphology of RNA-related granules such as Gems. CONCLUSIONS: Collectively, FUS/TLS depletion causes phenotypes possibly related to neuropsychiatric and neurodegenerative conditions, but distinct from ALS and ET, together with specific alterations in RNA metabolisms.


Amyotrophic Lateral Sclerosis/physiopathology , Anxiety/psychology , Behavior, Animal , RNA-Binding Protein FUS/genetics , RNA-Binding Proteins/genetics , Amyotrophic Lateral Sclerosis/genetics , Animals , Disease Models, Animal , Essential Tremor/genetics , Essential Tremor/physiopathology , Heterogeneous-Nuclear Ribonucleoprotein Group A-B/genetics , Homozygote , Hyperkinesis , Mice , Mice, Inbred C57BL , Mice, Knockout , Phenotype , RNA-Binding Protein FUS/deficiency , TATA-Binding Protein Associated Factors/genetics , Up-Regulation
15.
Hum Mol Genet ; 24(4): 1092-105, 2015 Feb 15.
Article En | MEDLINE | ID: mdl-25305080

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.


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
16.
Hum Mol Genet ; 24(3): 740-56, 2015 Feb 01.
Article En | MEDLINE | ID: mdl-25274774

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.


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
17.
Nat Commun ; 5: 5525, 2014 Nov 21.
Article En | MEDLINE | ID: mdl-25413837

Voltage-gated Na(+) channel ß-subunits are multifunctional molecules that modulate Na(+) channel activity and regulate cell adhesion, migration and neurite outgrowth. ß-subunits including ß4 are known to be highly concentrated in the nodes of Ranvier and axon initial segments in myelinated axons. Here we show diffuse ß4 localization in striatal projection fibres using transgenic mice that express fluorescent protein in those fibres. These axons are unmyelinated, forming large, inhibitory fibre bundles. Furthermore, we report ß4 dimer expression in the mouse brain, with high levels of ß4 dimers in the striatal projection fascicles, suggesting a specific role of ß4 in those fibres. Scn4b-deficient mice show a resurgent Na(+) current reduction, decreased repetitive firing frequency in medium spiny neurons and increased failure rates of inhibitory postsynaptic currents evoked with repetitive stimulation, indicating an in vivo channel regulatory role of ß4 in the striatum.


Corpus Striatum/metabolism , Ion Channel Gating/physiology , Nerve Fibers, Unmyelinated/metabolism , Voltage-Gated Sodium Channel beta-4 Subunit/genetics , Action Potentials/physiology , Animals , Huntingtin Protein , Huntington Disease/pathology , Male , Mice , Mice, Knockout , Nerve Tissue Proteins/genetics , Nuclear Proteins/genetics , Patch-Clamp Techniques , RNA Interference , RNA, Small Interfering , Ranvier's Nodes/metabolism
18.
PLoS One ; 9(4): e93891, 2014.
Article En | MEDLINE | ID: mdl-24705917

In polyglutamine (polyQ) diseases including Huntington's disease (HD), mutant proteins containing expanded polyQ stretch form aggregates in neurons. Genetic or RNAi screenings in yeast, C. elegans or Drosophila have identified multiple genes modifying polyQ aggregation, a few of which are confirmed effective in mammals. However, the overall molecular mechanism underlying polyQ protein aggregation in mammalian cells still remains obscure. We here perform RNAi screening in mouse neuro2a cells to identify mammalian modifiers for aggregation of mutant huntingtin, a causative protein of HD. By systematic cell transfection and automated cell image analysis, we screen ∼ 12000 shRNA clones and identify 111 shRNAs that either suppress or enhance mutant huntingtin aggregation, without altering its gene expression. Classification of the shRNA-targets suggests that genes with various cellular functions such as gene transcription and protein phosphorylation are involved in modifying the aggregation. Subsequent analysis suggests that, in addition to the aggregation-modifiers sensitive to proteasome inhibition, some of them, such as a transcription factor Tcf20, and kinases Csnk1d and Pik3c2a, are insensitive to it. As for Tcf20, which contains polyQ stretches at N-terminus, its binding to mutant huntingtin aggregates is observed in neuro2a cells and in HD model mouse neurons. Notably, except Pik3c2a, the rest of the modifiers identified here are novel. Thus, our first large-scale RNAi screening in mammalian system identifies previously undescribed genetic players that regulate mutant huntingtin aggregation by several, possibly mammalian-specific mechanisms.


Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Phosphatidylinositol 3-Kinases/genetics , Protein Aggregation, Pathological/genetics , RNA Interference , RNA, Small Interfering/genetics , Transcription Factors/genetics , Animals , High-Throughput Screening Assays , Huntingtin Protein , Mice , Neurons/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Protein Aggregation, Pathological/metabolism , Transcription Factors/metabolism
19.
Biochim Biophys Acta ; 1842(9): 1472-84, 2014 Sep.
Article En | MEDLINE | ID: mdl-24769000

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.


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
20.
Nat Commun ; 5: 3354, 2014 Feb 25.
Article En | MEDLINE | ID: mdl-24566496

Nuclear transcription factor-Y (NF-Y), a key regulator of cell-cycle progression, often loses its activity during differentiation into nonproliferative cells. In contrast, NF-Y is still active in mature, differentiated neurons, although its neuronal significance remains obscure. Here we show that conditional deletion of the subunit NF-YA in postmitotic mouse neurons induces progressive neurodegeneration with distinctive ubiquitin/p62 pathology; these proteins are not incorporated into filamentous inclusion but co-accumulated with insoluble membrane proteins broadly on endoplasmic reticulum (ER). The degeneration also accompanies drastic ER disorganization, that is, an aberrant increase in ribosome-free ER in the perinuclear region, without inducing ER stress response. We further perform chromatin immunoprecipitation and identify several NF-Y physiological targets including Grp94 potentially involved in ER disorganization. We propose that NF-Y is involved in a unique regulation mechanism of ER organization in mature neurons and its disruption causes previously undescribed novel neuropathology accompanying abnormal ubiquitin/p62 accumulation.


Adaptor Proteins, Signal Transducing/metabolism , CCAAT-Binding Factor/metabolism , Endoplasmic Reticulum/metabolism , Heat-Shock Proteins/metabolism , Neurodegenerative Diseases/metabolism , Ubiquitin/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , CCAAT-Binding Factor/genetics , Cell Line, Tumor , Endoplasmic Reticulum Stress/genetics , Endoplasmic Reticulum Stress/physiology , Female , HSP70 Heat-Shock Proteins/genetics , HSP70 Heat-Shock Proteins/metabolism , Heat-Shock Proteins/genetics , Male , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Neurodegenerative Diseases/genetics , Sequestosome-1 Protein , Ubiquitin/genetics
...