Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Sci Rep ; 10(1): 17995, 2020 10 22.
Article in English | MEDLINE | ID: mdl-33093587

ABSTRACT

Genetic variations of TREM2 have been implicated as a risk factor of Alzheimer's disease (AD). Recent studies suggest that the loss of TREM2 function compromises microglial responses to the accumulation of amyloid beta. Previously, we found that exon 3 of TREM2 is an alternative exon whose skipping leads to a reduction in full-length TREM2 protein by inducing nonsense-mediated mRNA decay. Here, we aimed to identify factors regulating TREM2 splicing. Using a panel of RNA-binding proteins, we found that exon 3 skipping of TREM2 was promoted by two paralogous proteins, CELF1 and CELF2, which were both linked previously with risk loci of AD. Although the overexpression of both CELF1 and CELF2 enhanced exon 3 skipping, only CELF2 reduced the expression of full-length TREM2 protein. Notably, the TREM2 ortholog in the green monkey, but not in the mouse, showed alternative splicing of exon 3 like human TREM2. Similarly, splicing regulation of exon 3 by CELF1/2 was found to be common to humans and monkeys. Using chimeric minigenes of human and mouse TREM2, we mapped a CELF-responsive sequence within intron 3 of human TREM2. Collectively, our results revealed a novel regulatory factor of TREM2 expression and highlighted a species-dependent difference of its regulation.


Subject(s)
Alternative Splicing , CELF Proteins/metabolism , Exons , Gene Expression Regulation , Membrane Glycoproteins/genetics , Nerve Tissue Proteins/metabolism , Receptors, Immunologic/genetics , Animals , CELF Proteins/genetics , HEK293 Cells , Humans , Membrane Glycoproteins/metabolism , Mice , Nerve Tissue Proteins/genetics , Receptors, Immunologic/metabolism , Species Specificity
2.
Sci Rep ; 6: 35236, 2016 10 14.
Article in English | MEDLINE | ID: mdl-27739513

ABSTRACT

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.


Subject(s)
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
3.
Acta Neuropathol Commun ; 3: 24, 2015 Apr 25.
Article in English | MEDLINE | ID: mdl-25907258

ABSTRACT

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.


Subject(s)
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
4.
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
5.
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
6.
Nucleic Acids Res ; 39(7): 2781-98, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21109527

ABSTRACT

TLS (translocated in liposarcoma), also known as FUS (fused in sarcoma), is an RNA/DNA-binding protein that plays regulatory roles in transcription, pre-mRNA splicing and mRNA transport. Mutations in TLS are responsible for familial amyotrophic lateral sclerosis (ALS) type 6. Furthermore, TLS-containing intracellular inclusions are found in polyglutamine diseases, sporadic ALS, non-SOD1 familial ALS and a subset of frontotemporal lobar degeneration, indicating a pathological significance of TLS in a wide variety of neurodegenerative diseases. Here, we identified TLS domains that determine intracellular localization of the murine TLS. Among them, PY-NLS located in the C-terminus is a strong determinant of intracellular localization as well as splicing regulation of an E1A-derived minigene. Disruption of PY-NLS promoted the formation of cytoplasmic granules that were partially overlapped with stress granules and P-bodies. Some of the ALS-linked mutations altered both intracellular localization and splicing regulation of TLS, while most mutations alone did not affect splicing regulation. However, phospho-mimetic substitution of Ser505 (or Ser513 in human) could enhance the effects of ALS mutations, highlighting interplay between post-translational modification and ALS-linked mutations. These results demonstrate that ALS-linked mutations can variably cause loss of nuclear functions of TLS depending on the degree of impairment in nuclear localization.


Subject(s)
Amyotrophic Lateral Sclerosis/genetics , Mutation , RNA Splicing , RNA-Binding Protein FUS/genetics , Adenovirus E1A Proteins/genetics , Adenovirus E1A Proteins/metabolism , Animals , Cell Line , Cytoplasmic Granules/chemistry , Green Fluorescent Proteins/genetics , Humans , Mice , RNA-Binding Protein FUS/analysis , Recombinant Fusion Proteins/analysis
7.
Nucleic Acids Res ; 37(19): 6477-90, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19720736

ABSTRACT

The expression and function of the skeletal muscle chloride channel CLCN1/ClC-1 is regulated by alternative splicing. Inclusion of the CLCN1 exon 7A is aberrantly elevated in myotonic dystrophy (DM), a genetic disorder caused by the expansion of a CTG or CCTG repeat. Increased exon 7A inclusion leads to a reduction in CLCN1 function, which can be causative of myotonia. Two RNA-binding protein families--muscleblind-like (MBNL) and CUG-BP and ETR-3-like factor (CELF) proteins--are thought to mediate the splicing misregulation in DM. Here, we have identified multiple factors that regulate the alternative splicing of a mouse Clcn1 minigene. The inclusion of exon 7A was repressed by MBNL proteins while promoted by an expanded CUG repeat or CELF4, but not by CUG-BP. Mutation analyses suggested that exon 7A and its flanking region mediate the effect of MBNL1, whereas another distinct region in intron 6 mediates that of CELF4. An exonic splicing enhancer essential for the inclusion of exon 7A was identified at the 5' end of this exon, which might be inhibited by MBNL1. Collectively, these results provide a mechanistic model for the regulation of Clcn1 splicing, and reveal novel regulatory properties of MBNL and CELF proteins.


Subject(s)
Alternative Splicing , Chloride Channels/genetics , RNA-Binding Proteins/metabolism , Animals , CELF Proteins , Chloride Channels/metabolism , DNA Repeat Expansion , Exons , Humans , Mice , Muscle, Skeletal/metabolism , RNA Splice Sites , RNA, Messenger/metabolism , RNA-Binding Proteins/antagonists & inhibitors
8.
Hum Mol Genet ; 17(20): 3223-35, 2008 Oct 15.
Article in English | MEDLINE | ID: mdl-18658163

ABSTRACT

Huntington's disease (HD) is a fatal neurodegenerative disorder. Despite a tremendous effort to develop therapeutic tools in several HD models, there is no effective cure at present. Acidosis has been observed previously in cellular and in in vivo models as well as in the brains of HD patients. Here we challenged HD models with amiloride (Ami) derivative benzamil (Ben), a chemical agent used to rescue acid-sensing ion channel (ASIC)-dependent acidotoxicity, to examine whether chronic acidosis is an important part of the HD pathomechanism and whether these drugs could be used as novel therapeutic agents. Ben markedly reduced the huntingtin-polyglutamine (htt-polyQ) aggregation in an inducible cellular system, and the therapeutic value of Ben was successfully recapitulated in the R6/2 animal model of HD. To reveal the mechanism of action, Ben was found to be able to alleviate the inhibition of the ubiquitin-proteasome system (UPS) activity, resulting in enhanced degradation of soluble htt-polyQ specifically in its pathological range. More importantly, we were able to demonstrate that blocking the expression of a specific isoform of ASIC (asic1a), one of the many molecular targets of Ben, led to an enhancement of UPS activity and this blockade also decreased htt-polyQ aggregation in the striatum of R6/2 mice. In conclusion, we believe that chemical compounds that target ASIC1a or pharmacological alleviation of UPS inhibition would be an effective and promising approach to combat HD and other polyQ-related disorders.


Subject(s)
Huntington Disease/drug therapy , Nerve Tissue Proteins/antagonists & inhibitors , Acid Sensing Ion Channels , Adult , Amiloride/analogs & derivatives , Amiloride/pharmacology , Animals , Cells, Cultured , Disease Models, Animal , Female , Humans , Huntington Disease/genetics , Huntington Disease/metabolism , In Vitro Techniques , Male , Mice , Mice, Transgenic , Middle Aged , Mutation , Nerve Tissue Proteins/genetics , Peptides/chemistry , Peptides/genetics , RNA Interference , Rats , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Serotonin Plasma Membrane Transport Proteins/chemistry , Serotonin Plasma Membrane Transport Proteins/genetics , Serotonin Plasma Membrane Transport Proteins/metabolism , Sodium Channel Blockers/pharmacology , Sodium Channels/genetics , Solubility
9.
EMBO J ; 27(6): 827-39, 2008 Mar 19.
Article in English | MEDLINE | ID: mdl-18288205

ABSTRACT

In Huntington's disease (HD), mutant Huntingtin, which contains expanded polyglutamine stretches, forms nuclear aggregates in neurons. The interactions of several transcriptional factors with mutant Huntingtin, as well as altered expression of many genes in HD models, imply the involvement of transcriptional dysregulation in the HD pathological process. The precise mechanism remains obscure, however. Here, we show that mutant Huntingtin aggregates interact with the components of the NF-Y transcriptional factor in vitro and in HD model mouse brain. An electrophoretic mobility shift assay using HD model mouse brain lysates showed reduction in NF-Y binding to the promoter region of HSP70, one of the NF-Y targets. RT-PCR analysis revealed reduced HSP70 expression in these brains. We further clarified the importance of NF-Y for HSP70 transcription in cultured neurons. These data indicate that mutant Huntingtin sequesters NF-Y, leading to the reduction of HSP70 gene expression in HD model mice brain. Because suppressive roles of HSP70 on the HD pathological process have been shown in several HD models, NF-Y could be an important target of mutant Huntingtin.


Subject(s)
CCAAT-Binding Factor/metabolism , HSP70 Heat-Shock Proteins/antagonists & inhibitors , HSP70 Heat-Shock Proteins/genetics , Mutation , Nerve Tissue Proteins/genetics , Nuclear Proteins/genetics , Animals , CCAAT-Binding Factor/antagonists & inhibitors , Cell Line, Tumor , Disease Models, Animal , HSP70 Heat-Shock Proteins/biosynthesis , Huntingtin Protein , Huntington Disease/genetics , Huntington Disease/metabolism , Male , Mice , Mice, Transgenic , Nerve Tissue Proteins/physiology , Nuclear Proteins/physiology , Protein Binding/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...