Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Mol Neurodegener ; 16(1): 61, 2021 09 06.
Article in English | MEDLINE | ID: mdl-34488813

ABSTRACT

Mutations in FUS, an RNA-binding protein involved in multiple steps of RNA metabolism, are associated with the most severe forms of amyotrophic lateral sclerosis (ALS). Accumulation of cytoplasmic FUS is likely to be a major culprit in the toxicity of FUS mutations. Thus, preventing cytoplasmic mislocalization of the FUS protein may represent a valuable therapeutic strategy. FUS binds to its own pre-mRNA creating an autoregulatory loop efficiently buffering FUS excess through multiple proposed mechanisms including retention of introns 6 and/or 7. Here, we introduced a wild-type FUS gene allele, retaining all intronic sequences, in mice whose heterozygous or homozygous expression of a cytoplasmically retained FUS protein (Fus∆NLS) was previously shown to provoke ALS-like disease or postnatal lethality, respectively. Wild-type FUS completely rescued the early lethality caused by the two Fus∆NLS alleles, and improved the age-dependent motor deficits and reduced lifespan caused by heterozygous expression of mutant FUS∆NLS. Mechanistically, wild-type FUS decreased the load of cytoplasmic FUS, increased retention of introns 6 and 7 in the endogenous mouse Fus mRNA, and decreased expression of the mutant mRNA. Thus, the wild-type FUS allele activates the homeostatic autoregulatory loop, maintaining constant FUS levels and decreasing the mutant protein in the cytoplasm. These results provide proof of concept that an autoregulatory competent wild-type FUS expression could protect against this devastating, currently intractable, neurodegenerative disease.


Subject(s)
Amyotrophic Lateral Sclerosis/therapy , Disease Models, Animal , Gene Expression Regulation/genetics , RNA-Binding Protein FUS/physiology , Alleles , Amyotrophic Lateral Sclerosis/genetics , Animals , Cytoplasm/metabolism , Frontotemporal Dementia/genetics , Genes, Lethal , Genetic Complementation Test , Humans , Introns/genetics , Mice , Mice, Transgenic , Mutation , Protein Binding , RNA Precursors/metabolism , RNA-Binding Protein FUS/deficiency , RNA-Binding Protein FUS/genetics , Recombinant Proteins/metabolism , Transgenes
2.
Prog Mol Biol Transl Sci ; 172: 157-202, 2020.
Article in English | MEDLINE | ID: mdl-32620242

ABSTRACT

Motor neuron diseases (MNDs) are a wide group of neurodegenerative disorders characterized by the degeneration of a specific neuronal type located in the central nervous system, the motor neuron (MN). There are two main types of MNs, spinal and cortical MNs and depending on the type of MND, one or both types are affected. Cortical MNs innervate spinal MNs and these control a variety of cellular targets, being skeletal muscle their main one which is also affected in MNDs. A correct functionality of autophagy is necessary for the survival of all cellular types and it is particularly crucial for neurons, given their postmitotic and highly specialized nature. Numerous studies have identified alterations of autophagy activity in multiple MNDs. The scientific community has been particularly prolific in reporting the role that autophagy plays in the most common adult MND, amyotrophic lateral sclerosis, although many studies have started to identify physiological and pathological functions of this catabolic system in other MNDs, such as spinal muscular atrophy and spinal and bulbar muscular atrophy. The degradation of selective cargo by autophagy and how this process is altered upon the presence of MND-causing mutations is currently also a matter of intense investigation, particularly regarding the selective autophagic clearance of mitochondria. Thorough reviews on this field have been recently published. This chapter will cover the current knowledge on the functionality of autophagy and lysosomal homeostasis in the main MNDs and other autophagy-related topics in the MND field that have risen special interest in the research community.


Subject(s)
Autophagy , Motor Neuron Disease/pathology , Adult , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/pathology , Animals , Autophagy/drug effects , Autophagy/physiology , Autophagy-Related Proteins/genetics , Autophagy-Related Proteins/physiology , C9orf72 Protein/deficiency , C9orf72 Protein/genetics , C9orf72 Protein/physiology , DNA Repeat Expansion , Disease Models, Animal , Endocytosis , Humans , Mice, Transgenic , Motor Neuron Disease/genetics , Muscular Atrophy, Spinal/genetics , Muscular Atrophy, Spinal/pathology , Mutation , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/pathology , Organelles , RNA-Binding Protein FUS/deficiency , RNA-Binding Protein FUS/genetics , RNA-Binding Protein FUS/physiology , TDP-43 Proteinopathies/genetics , TDP-43 Proteinopathies/pathology
3.
Glia ; 68(10): 2040-2056, 2020 10.
Article in English | MEDLINE | ID: mdl-32187401

ABSTRACT

Fused in sarcoma (FUS) is a predominantly nuclear multifunctional RNA/DNA-binding protein that regulates multiple aspects of gene expression. FUS mutations are associated with familial amyotrophic lateral sclerosis (fALS) and frontotemporal lobe degeneration (FTLD) in humans. At the molecular level, the mutated FUS protein is reduced in the nucleus but accumulates in cytoplasmic granules. Oligodendrocytes (OL) carrying clinically relevant FUS mutations contribute to non-cell autonomous motor neuron disease progression, consistent with an extrinsic mechanism of disease mediated by OL. Knocking out FUS globally or in neurons lead to behavioral abnormalities that are similar to those present in FTLD. In this study, we sought to investigate whether an extrinsic mechanism mediated by loss of FUS function in OL contributes to the behavioral phenotype. We have generated a novel conditional knockout (cKO) in which Fus is selectively depleted in OL (FusOL cKO). The FusOL cKO mice show increased novelty-induced motor activity and enhanced exploratory behavior, which are reminiscent of some manifestations of FTLD. The phenotypes are associated with greater myelin thickness, higher number of myelinated small diameter axons without an increase in the number of mature OL. The expression of the rate-limiting enzyme of cholesterol biosynthesis (HMGCR) is increased in white matter tracts of the FusOL cKO and results in higher cholesterol content. In addition, phosphorylation of Akt, an important regulator of myelination is increased in the FusOL cKO. Collectively, this work has uncovered a novel role of oligodendrocytic Fus in regulating myelin deposition through activation of Akt and cholesterol biosynthesis.


Subject(s)
Cholesterol/metabolism , Hyperkinesis/metabolism , Myelin Sheath/metabolism , Oligodendroglia/metabolism , Proto-Oncogene Proteins c-akt/metabolism , RNA-Binding Protein FUS/deficiency , Animals , Cholesterol/genetics , Hyperkinesis/genetics , Maze Learning/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Myelin Sheath/genetics , Proto-Oncogene Proteins c-akt/genetics , RNA-Binding Protein FUS/genetics
4.
Nucleic Acids Res ; 46(22): 11939-11951, 2018 12 14.
Article in English | MEDLINE | ID: mdl-30398641

ABSTRACT

Understanding the molecular pathways disrupted in motor neuron diseases is urgently needed. Here, we employed CRISPR knockout (KO) to investigate the functions of four ALS-causative RNA/DNA binding proteins (FUS, EWSR1, TAF15 and MATR3) within the RNAP II/U1 snRNP machinery. We found that each of these structurally related proteins has distinct roles with FUS KO resulting in loss of U1 snRNP and the SMN complex, EWSR1 KO causing dissociation of the tRNA ligase complex, and TAF15 KO resulting in loss of transcription factors P-TEFb and TFIIF. However, all four ALS-causative proteins are required for association of the ASC-1 transcriptional co-activator complex with the RNAP II/U1 snRNP machinery. Remarkably, mutations in the ASC-1 complex are known to cause a severe form of Spinal Muscular Atrophy (SMA), and we show that an SMA-causative mutation in an ASC-1 component or an ALS-causative mutation in FUS disrupts association between the ASC-1 complex and the RNAP II/U1 snRNP machinery. We conclude that ALS and SMA are more intimately tied to one another than previously thought, being linked via the ASC-1 complex.


Subject(s)
Amyotrophic Lateral Sclerosis/genetics , Muscular Atrophy, Spinal/genetics , Nuclear Matrix-Associated Proteins/genetics , RNA-Binding Protein EWS/genetics , RNA-Binding Protein FUS/genetics , RNA-Binding Proteins/genetics , TATA-Binding Protein Associated Factors/genetics , Amyotrophic Lateral Sclerosis/metabolism , Amyotrophic Lateral Sclerosis/pathology , CRISPR-Cas Systems , Carrier Proteins/genetics , Carrier Proteins/metabolism , DNA Helicases/genetics , DNA Helicases/metabolism , Gene Editing , Gene Expression Regulation , Gene Knockout Techniques , Humans , Muscular Atrophy, Spinal/metabolism , Muscular Atrophy, Spinal/pathology , Nuclear Matrix-Associated Proteins/deficiency , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Positive Transcriptional Elongation Factor B/genetics , Positive Transcriptional Elongation Factor B/metabolism , RNA Polymerase II/genetics , RNA Polymerase II/metabolism , RNA, Transfer/genetics , RNA, Transfer/metabolism , RNA-Binding Protein EWS/deficiency , RNA-Binding Protein FUS/deficiency , Ribonucleoprotein, U1 Small Nuclear/genetics , Ribonucleoprotein, U1 Small Nuclear/metabolism , Spliceosomes/chemistry , Spliceosomes/metabolism , TATA-Binding Protein Associated Factors/deficiency , Transcription Factors, TFII/genetics , Transcription Factors, TFII/metabolism
5.
Biochim Biophys Acta ; 1849(12): 1398-410, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26514432

ABSTRACT

The full definition of the physiological RNA targets regulated by TDP-43 and FUS RNA-binding proteins (RBPs) represents an important issue in understanding the pathogenic mechanisms associated to these two proteins in amyotrophic lateral sclerosis and frontotemporal dementia. In the last few years several high-throughput screenings have generated a plethora of data, which are difficult to compare due to the different experimental designs and models explored. In this study by using the Affymetrix Exon Arrays, we were able to assess and compare the effects of both TDP-43 and FUS loss-of-function on the whole transcriptome using the same human neuronal SK-N-BE cell model. We showed that TDP-43 and FUS depletion induces splicing and gene expression changes mainly distinct for the two RBPs, although they may regulate common pathways, including neuron differentiation and cytoskeleton organization as evidenced by functional annotation analysis. In particular, TDP-43 and FUS were found to regulate splicing and expression of genes related to neuronal (SEPT6, SULT4A1, TNIK) and RNA metabolism (DICER, ELAVL3/HuC, POLDIP3). Our extended analysis at protein level revealed that these changes have also impact on the protein isoform ratio and content, not always in a direct correlation with transcriptomic data. Contrarily to a loss-of-function mechanism, we showed that mutant TDP-43 proteins maintained their splicing activity in human ALS fibroblasts and experimental cell lines. Our findings further contribute to define the biological functions of these two RBPs in physiological and disease state, strongly encouraging the evaluation of the identified transcriptomic changes at protein level in neuronal experimental models.


Subject(s)
DNA-Binding Proteins/physiology , Gene Expression Regulation, Neoplastic/genetics , Neoplasm Proteins/physiology , Neurons/metabolism , Proteome , RNA Precursors/genetics , RNA, Messenger/genetics , RNA, Neoplasm/genetics , RNA-Binding Protein FUS/physiology , Transcriptome , Alternative Splicing , Amino Acid Sequence , Animals , Base Sequence , Cell Line, Tumor , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Exons/genetics , Fibroblasts/metabolism , Humans , Mice , Mice, Inbred BALB C , Models, Genetic , Molecular Sequence Data , Neoplasm Proteins/deficiency , Neoplasm Proteins/genetics , Neuroblastoma/pathology , Protein Isoforms/metabolism , RNA Interference , RNA Precursors/metabolism , RNA, Messenger/metabolism , RNA, Neoplasm/metabolism , RNA, Small Interfering/genetics , RNA-Binding Protein FUS/deficiency , RNA-Binding Protein FUS/genetics , Sequence Alignment , Sequence Homology
6.
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
7.
Cell Death Dis ; 5: e1572, 2014 Dec 11.
Article in English | MEDLINE | ID: mdl-25501833

ABSTRACT

Fused in sarcoma/translocated in liposarcoma (FUS/TLS or FUS) is a multifunctional RNA/DNA-binding protein that is pathologically associated with cancer and neurodegeneration. To gain insight into the vital functions of FUS and how a loss of FUS function impacts cellular homeostasis, FUS expression was reduced in different cellular models through RNA interference. Our results show that a loss of FUS expression severely impairs cellular proliferation and leads to an increase in phosphorylated histone H3, a marker of mitotic arrest. A quantitative proteomics analysis performed on cells undergoing various degrees of FUS knockdown revealed protein expression changes for known RNA targets of FUS, consistent with a loss of FUS function with respect to RNA processing. Proteins that changed in expression as a function of FUS knockdown were associated with multiple processes, some of which influence cell proliferation including cell cycle regulation, cytoskeletal organization, oxidative stress and energy homeostasis. FUS knockdown also correlated with increased expression of the closely related protein EWS (Ewing's sarcoma). We demonstrate that the maladaptive phenotype resulting from FUS knockdown is reversible and can be rescued by re-expression of FUS or partially rescued by the small-molecule rolipram. These results provide insight into the pathways and processes that are regulated by FUS, as well as the cellular consequences for a loss of FUS function.


Subject(s)
Cell Proliferation , Cells/cytology , RNA-Binding Protein FUS/deficiency , Cell Line , Cells/metabolism , Gene Knockdown Techniques , Histones/metabolism , Humans , M Phase Cell Cycle Checkpoints , Phosphorylation , RNA Interference , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , RNA-Binding Protein FUS/genetics
8.
PLoS One ; 8(4): e60788, 2013.
Article in English | MEDLINE | ID: mdl-23577159

ABSTRACT

Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disease resulting in severe muscle weakness and eventual death by respiratory failure. Although little is known about its pathogenesis, mutations in fused in sarcoma/translated in liposarcoma (FUS) are causative for familial ALS. FUS is a multifunctional protein that is involved in many aspects of RNA processing. To elucidate the role of FUS in ALS, we overexpressed wild-type and two mutant forms of FUS in HEK-293T cells, as well as knocked-down FUS expression. This was followed by RNA-Seq to identify genes which displayed differential expression or altered splicing patterns. Pathway analysis revealed that overexpression of wild-type FUS regulates ribosomal genes, whereas knock-down of FUS additionally affects expression of spliceosome related genes. Furthermore, cells expressing mutant FUS displayed global transcription patterns more similar to cells overexpressing wild-type FUS than to the knock-down condition. This observation suggests that FUS mutants do not contribute to the pathogenesis of ALS through a loss-of-function. Finally, our results demonstrate that the R521G and R522G mutations display differences in their influence on transcription and splicing. Taken together, these results provide additional insights into the function of FUS and how mutations contribute to the development of ALS.


Subject(s)
Amyotrophic Lateral Sclerosis/genetics , Mutation , RNA-Binding Protein FUS/genetics , Sequence Analysis, RNA , Exons/genetics , HEK293 Cells , Humans , Introns/genetics , RNA, Small Interfering/genetics , RNA-Binding Protein FUS/deficiency
9.
Nat Neurosci ; 15(11): 1488-97, 2012 Nov.
Article in English | MEDLINE | ID: mdl-23023293

ABSTRACT

FUS/TLS (fused in sarcoma/translocated in liposarcoma) and TDP-43 are integrally involved in amyotrophic lateral sclerosis (ALS) and frontotemporal dementia. We found that FUS/TLS binds to RNAs from >5,500 genes in mouse and human brain, primarily through a GUGGU-binding motif. We identified a sawtooth-like binding pattern, consistent with co-transcriptional deposition of FUS/TLS. Depletion of FUS/TLS from the adult nervous system altered the levels or splicing of >950 mRNAs, most of which are distinct from RNAs dependent on TDP-43. Abundance of only 45 RNAs was reduced after depletion of either TDP-43 or FUS/TLS from mouse brain, but among these were mRNAs that were transcribed from genes with exceptionally long introns and that encode proteins that are essential for neuronal integrity. Expression levels of a subset of these were lowered after TDP-43 or FUS/TLS depletion in stem cell-derived human neurons and in TDP-43 aggregate-containing motor neurons in sporadic ALS, supporting a common loss-of-function pathway as one component underlying motor neuron death from misregulation of TDP-43 or FUS/TLS.


Subject(s)
Amyotrophic Lateral Sclerosis/metabolism , DNA-Binding Proteins/metabolism , Frontotemporal Dementia/metabolism , RNA Precursors/metabolism , RNA, Messenger/metabolism , RNA-Binding Protein FUS/metabolism , Adaptor Proteins, Signal Transducing , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/pathology , Animals , Autophagy-Related Proteins , Brain/metabolism , Brain/pathology , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Line, Transformed , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Excitatory Amino Acid Transporter 2/genetics , Excitatory Amino Acid Transporter 2/metabolism , Female , Frontotemporal Dementia/genetics , Frontotemporal Dementia/pathology , Gene Expression Profiling , Gene Expression Regulation/genetics , Histone-Lysine N-Methyltransferase/metabolism , Humans , Immunoprecipitation , Kv Channel-Interacting Proteins/metabolism , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Motor Neurons/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neural Cell Adhesion Molecules/metabolism , Neural Stem Cells/metabolism , Neurofilament Proteins/metabolism , Oligonucleotide Array Sequence Analysis , Protein Binding/genetics , Protein Structure, Tertiary/genetics , RNA Precursors/genetics , RNA Splicing/genetics , RNA, Messenger/genetics , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , RNA-Binding Protein FUS/deficiency , RNA-Binding Protein FUS/genetics , Shal Potassium Channels/metabolism , Spinal Cord/metabolism , Ubiquitin-Protein Ligases/metabolism , tau Proteins/genetics , tau Proteins/metabolism
10.
Rev Neurol (Paris) ; 166(8-9): 683-98, 2010.
Article in French | MEDLINE | ID: mdl-20472259

ABSTRACT

This paper, written by French amyotrophic lateral sclerosis (ALS) center experts, presents an update of recent advances in fundamental, epidemiological and clinical research in ALS based on a review of the literature between September 2008 and November 2009. Among other pathophysiological mechanisms, the role of stress of the endoplasmic reticulum and the importance of energetic metabolic disturbances have been underscored. In the field of genetics, research has been advanced through the identification of mutations of the gene FUsed in Sarcoma/Translated in LipoSarcoma (FUS/TLS) in individuals with familial and sporadic ALS. This gene is involved in the regulation of transcription, splicing and RNA transport, and has functional homology to another ALS gene, TARDBP, which suggests that a common mechanism may underlie motor neuron degeneration. A report showed that mice expressing a mutant form of human TDP-43 develop a progressive and fatal neurodegenerative disease reminiscent of both ALS and frontotemporal lobar degeneration with ubiquitin aggregates (FTLD-U), providing a new animal model that may help to better understand the pathophysiology and test new therapeutics. Beside genetic studies, several epidemiologic studies have investigated the role of environmental factors. A recent study suggests that smoking is a risk factor for developing ALS and it is hypothesized that this could occur through lipid peroxidation via formaldehyde exposure. From a neuroprotective perspective, trials with IGF-1, sodium valproate, coenzyme Q or glatiramer acetate have failed to demonstrate any beneficial effect. A study published in 2008 argued that lithium may have a neuroprotective effect in ALS mice and also in patients. However, two preclinical studies failed to replicate the neuroprotective effect of lithium in ALS mice. Therapeutic trials have been performed or are currently ongoing in Europe and North America. Their results have not yet been published.


Subject(s)
Amyotrophic Lateral Sclerosis , Amyotrophic Lateral Sclerosis/drug therapy , Amyotrophic Lateral Sclerosis/epidemiology , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/physiopathology , Amyotrophic Lateral Sclerosis/psychology , Animals , Biomarkers , Clinical Trials as Topic , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , DNA-Binding Proteins/physiology , Disease Models, Animal , Drug Evaluation, Preclinical , Environmental Exposure , Humans , Malnutrition/etiology , Malnutrition/therapy , Mice , Mice, Transgenic , Muscle, Skeletal/metabolism , Neuroprotective Agents/therapeutic use , RNA-Binding Protein FUS/deficiency , RNA-Binding Protein FUS/genetics , RNA-Binding Protein FUS/physiology , Risk Factors , Superoxide Dismutase/deficiency , Superoxide Dismutase/genetics , Superoxide Dismutase/physiology , Superoxide Dismutase-1
SELECTION OF CITATIONS
SEARCH DETAIL
...