Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 50
1.
Mol Neurodegener ; 18(1): 5, 2023 01 18.
Article En | MEDLINE | ID: mdl-36653804

BACKGROUND: Astrocytes play a crucial, yet not fully elucidated role in the selective motor neuron pathology in amyotrophic lateral sclerosis (ALS). Among other responsibilities, astrocytes provide important neuronal homeostatic support, however this function is highly compromised in ALS. The establishment of fully human coculture systems can be used to further study the underlying mechanisms of the dysfunctional intercellular interplay, and has the potential to provide a platform for revealing novel therapeutic entry points. METHODS: In this study, we characterised human induced pluripotent stem cell (hiPSC)-derived astrocytes from FUS-ALS patients, and incorporated these cells into a human motor unit microfluidics model to investigate the astrocytic effect on hiPSC-derived motor neuron network and functional neuromuscular junctions (NMJs) using immunocytochemistry and live-cell recordings. FUS-ALS cocultures were systematically compared to their CRISPR-Cas9 gene-edited isogenic control systems. RESULTS: We observed a dysregulation of astrocyte homeostasis, which resulted in a FUS-ALS-mediated increase in reactivity and secretion of inflammatory cytokines. Upon coculture with motor neurons and myotubes, we detected a cytotoxic effect on motor neuron-neurite outgrowth, NMJ formation and functionality, which was improved or fully rescued by isogenic control astrocytes. We demonstrate that ALS astrocytes have both a gain-of-toxicity and loss-of-support function involving the WNT/ß-catenin pathway, ultimately contributing to the disruption of motor neuron homeostasis, intercellular networks and NMJs. CONCLUSIONS: Our findings shine light on a complex, yet highly important role of astrocytes in ALS, and provides further insight in to their pathological mechanisms.


Amyotrophic Lateral Sclerosis , Induced Pluripotent Stem Cells , Humans , Amyotrophic Lateral Sclerosis/metabolism , Astrocytes/metabolism , Induced Pluripotent Stem Cells/metabolism , Motor Neurons/metabolism , Neuromuscular Junction , RNA-Binding Protein FUS/physiology
2.
Biochemistry ; 60(43): 3213-3222, 2021 11 02.
Article En | MEDLINE | ID: mdl-34648275

Glycine-rich regions feature prominently in intrinsically disordered regions (IDRs) of proteins that drive phase separation and the regulated formation of membraneless biomolecular condensates. Interestingly, the Gly-rich IDRs seldom feature poly-Gly tracts. The protein fused in sarcoma (FUS) is an exception. This protein includes two 10-residue poly-Gly tracts within the prion-like domain (PLD) and at the interface between the PLD and the RNA binding domain. Poly-Gly tracts are known to be highly insoluble, being potent drivers of self-assembly into solid-like fibrils. Given that the internal concentrations of FUS and FUS-like molecules cross the high micromolar and even millimolar range within condensates, we reasoned that the intrinsic insolubility of poly-Gly tracts might be germane to emergent fluid-to-solid transitions within condensates. To assess this possibility, we characterized the concentration-dependent self-assembly for three non-overlapping 25-residue Gly-rich peptides derived from FUS. Two of the three peptides feature 10-residue poly-Gly tracts. These peptides form either long fibrils based on twisted ribbon-like structures or self-supporting gels based on physical cross-links of fibrils. Conversely, the peptide with similar Gly contents but lacking a poly-Gly tract does not form fibrils or gels. Instead, it remains soluble across a wide range of concentrations. Our findings highlight the ability of poly-Gly tracts within IDRs that drive phase separation to undergo self-assembly. We propose that these tracts are likely to contribute to nucleation of fibrillar solids within dense condensates formed by FUS.


Glycine/metabolism , Peptides/chemistry , RNA-Binding Protein FUS/metabolism , Biomolecular Condensates/metabolism , Intrinsically Disordered Proteins/metabolism , Microscopy, Electron, Transmission/methods , Peptides/metabolism , Protein Aggregates/genetics , Protein Aggregates/physiology , Protein Domains/physiology , RNA-Binding Protein FUS/genetics , RNA-Binding Protein FUS/physiology , Spectroscopy, Fourier Transform Infrared/methods
3.
Int J Mol Sci ; 22(20)2021 Oct 18.
Article En | MEDLINE | ID: mdl-34681883

Amyotrophic lateral sclerosis (ALS) is an incurable and fatal neurodegenerative disorder of the motor system. While the etiology is still incompletely understood, defects in metabolism act as a major contributor to the disease progression. Recently, histone deacetylase (HDAC) inhibition using ACY-738 has been shown to restore metabolic alterations in the spinal cord of a FUS mouse model of ALS, which was accompanied by a beneficial effect on the motor phenotype and survival. In this study, we investigated the specific effects of HDAC inhibition on lipid metabolism using untargeted lipidomic analysis combined with transcriptomic analysis in the spinal cord of FUS mice. We discovered that symptomatic FUS mice recapitulate lipid alterations found in ALS patients and in the SOD1 mouse model. Glycerophospholipids, sphingolipids, and cholesterol esters were most affected. Strikingly, HDAC inhibition mitigated lipid homeostasis defects by selectively targeting glycerophospholipid metabolism and reducing cholesteryl esters accumulation. Therefore, our data suggest that HDAC inhibition is a potential new therapeutic strategy to modulate lipid metabolism defects in ALS and potentially other neurodegenerative diseases.


Amyotrophic Lateral Sclerosis/metabolism , Disease Models, Animal , Histone Deacetylase Inhibitors/pharmacology , Lipids/analysis , RNA-Binding Protein FUS/physiology , Transcriptome/drug effects , Amyotrophic Lateral Sclerosis/drug therapy , Amyotrophic Lateral Sclerosis/pathology , Animals , Female , Hydroxamic Acids/pharmacology , Lipidomics , Male , Mice , Pyrimidines/pharmacology
4.
Mol Neurodegener ; 16(1): 61, 2021 09 06.
Article En | MEDLINE | ID: mdl-34488813

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.


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
5.
J Neurochem ; 157(3): 752-763, 2021 05.
Article En | MEDLINE | ID: mdl-33354770

Fused in sarcoma (FUS) is a ubiquitously expressed RNA/DNA-binding protein that plays different roles in the cell. FUS pathology has been reported in neurodegenerative diseases amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD). Mutations in FUS have also been linked to a subset of familial ALS. FUS is mainly localized in the nucleus although it shuttles between the nucleus and the cytoplasm. ALS-linked mutations cause the accumulation of the FUS protein in cytoplasm where it forms stress granule-like inclusions. The protein- and RNA-containing inclusions are reported to be positive of autophagosome markers and degraded by the autophagy pathway. However, the role of FUS in the autophagy pathway remains to be better understood. Using immunoblot and confocal imaging techniques in this study, we found that FUS knockout (KO) cells showed a decreased basal autophagy level. Rapamycin and bafilomycin A1 treatment showed that FUS KO cells were not able to initiate autophagy as efficiently as wild-type cells, suggesting that the autophagosome formation is affected in the absence of FUS. Moreover, using immunoblot and quantitative PCR techniques, we found that the mRNA and protein levels of the genes critical in the initial steps of the autophagy pathway (FIP200, ATG16L1 and ATG12) were significantly lower in FUS KO cells. Re-expressing FUS in the KO cells restored the expression of FIP200 and ATG16L1. Our findings demonstrate a novel role of FUS in the autophagy pathway, that is, regulating the transcription of genes involved in early stages of autophagy such as the initiation and elongation of autophagosomes.


Autophagosomes/genetics , Autophagosomes/physiology , Autophagy/genetics , Autophagy/physiology , RNA-Binding Protein FUS/genetics , RNA-Binding Protein FUS/physiology , Animals , Autophagosomes/drug effects , Autophagy/drug effects , Autophagy-Related Proteins/genetics , Autophagy-Related Proteins/physiology , Cell Line , Gene Expression Regulation , Gene Knockout Techniques , Macrolides/pharmacology , Mice , Proteasome Endopeptidase Complex , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Signal Transduction/genetics , Sirolimus/pharmacology
6.
Mol Biol Cell ; 31(23): 2522-2536, 2020 11 01.
Article En | MEDLINE | ID: mdl-32877292

Fused in Sarcoma (FUS) is a ubiquitously expressed protein that can phase-separate from nucleoplasm and cytoplasm into distinct liquid-droplet structures. It is predominantly nuclear and most of its functions are related to RNA and DNA metabolism. Excessive persistence of FUS within cytoplasmic phase-separated assemblies is implicated in the diseases amyotrophic lateral sclerosis and frontotemporal dementia. Phosphorylation of FUS's prion-like domain (PrLD) by nuclear phosphatidylinositol 3-kinase-related kinase (PIKK)-family kinases following DNA damage was previously shown to alter FUS's liquid-phase and solid-phase transitions in cell models and in vitro. However, proteomic data suggest that FUS's PrLD is phosphorylated at numerous additional sites, and it is unknown if other non-PIKK and nonnuclear kinases might be influencing FUS's phase transitions. Here we evaluate disease mutations and stress conditions that increase FUS accumulation into cytoplasmic phase-separated structures. We observed that cytoplasmic liquid-phase structures contain FUS phosphorylated at novel sites, which occurred independent of PIKK-family kinases. We engineered phosphomimetic substitutions within FUS's PrLD and observed that mimicking a few phosphorylation sites strongly inhibited FUS solid-phase aggregation, while minimally altering liquid-phase condensation. These effects occurred independent of the exact location of the phosphomimetic substitutions, suggesting that modulation of PrLD phosphorylation may offer therapeutic strategies that are specific for solid-phase aggregation observed in disease.


Phase Transition/drug effects , Prions/metabolism , RNA-Binding Protein FUS/metabolism , Cell Nucleus/metabolism , Cytoplasm/metabolism , DNA Damage , Humans , Mutation , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Prions/genetics , Protein Aggregation, Pathological , Protein Processing, Post-Translational , Proteomics , RNA-Binding Protein FUS/physiology
7.
Int J Mol Sci ; 21(19)2020 Sep 24.
Article En | MEDLINE | ID: mdl-32987654

The fused in sarcoma (FUS) protein combines prion-like properties with a multifunctional DNA/RNA-binding domain and has functions spanning the regulation of RNA metabolism, including transcription, pre-mRNA splicing, mRNA transport and translation. In addition to its roles in RNA metabolism, FUS is implicated in the maintenance of DNA integrity. In this review, we examine the participation of FUS in major DNA repair pathways, focusing on DNA repair associated with poly(ADP-ribosyl)ation events and on how the interaction of FUS with poly(ADP-ribose) may orchestrate transient compartmentalisation of DNA strand breaks. Unravelling how prion-like RNA-binding proteins control DNA repair pathways will deepen our understanding of the pathogenesis of some neurological diseases and cancer as well as provide the basis for the development of relevant innovative therapeutic technologies. This knowledge may also extend the range of applications of poly(ADP-ribose) polymerase inhibitors to the treatment of neurodegenerative diseases related to RNA-binding proteins in the cell, e.g., amyotrophic lateral sclerosis and frontotemporal lobar degeneration.


DNA Repair , Poly (ADP-Ribose) Polymerase-1/metabolism , RNA-Binding Protein FUS/physiology , Animals , Humans , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology
8.
Prog Mol Biol Transl Sci ; 172: 157-202, 2020.
Article En | MEDLINE | ID: mdl-32620242

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.


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
9.
Nat Neurosci ; 22(11): 1793-1805, 2019 11.
Article En | MEDLINE | ID: mdl-31591561

Neuromuscular junction (NMJ) disruption is an early pathogenic event in amyotrophic lateral sclerosis (ALS). Yet, direct links between NMJ pathways and ALS-associated genes such as FUS, whose heterozygous mutations cause aggressive forms of ALS, remain elusive. In a knock-in Fus-ALS mouse model, we identified postsynaptic NMJ defects in newborn homozygous mutants that were attributable to mutant FUS toxicity in skeletal muscle. Adult heterozygous knock-in mice displayed smaller neuromuscular endplates that denervated before motor neuron loss, which is consistent with 'dying-back' neuronopathy. FUS was enriched in subsynaptic myonuclei, and this innervation-dependent enrichment was distorted in FUS-ALS. Mechanistically, FUS collaborates with the ETS transcription factor ERM to stimulate transcription of acetylcholine receptor genes. Co-cultures of induced pluripotent stem cell-derived motor neurons and myotubes from patients with FUS-ALS revealed endplate maturation defects due to intrinsic FUS toxicity in both motor neurons and myotubes. Thus, FUS regulates acetylcholine receptor gene expression in subsynaptic myonuclei, and muscle-intrinsic toxicity of ALS mutant FUS may contribute to dying-back motor neuronopathy.


Amyotrophic Lateral Sclerosis/physiopathology , Gene Expression Regulation/physiology , Nerve Degeneration/physiopathology , Neuromuscular Junction/metabolism , RNA-Binding Protein FUS/physiology , Adult , Amyotrophic Lateral Sclerosis/pathology , Animals , Cells, Cultured , Female , Gene Knock-In Techniques , Humans , Male , Mice , Mice, Knockout , Motor Neurons/pathology , Muscle Fibers, Skeletal/pathology , Neuromuscular Junction/pathology , RNA-Binding Protein FUS/genetics , RNA-Binding Protein FUS/metabolism , Receptors, Cholinergic/metabolism , Young Adult
10.
Med Sci Monit ; 24: 8750-8757, 2018 Dec 03.
Article En | MEDLINE | ID: mdl-30507891

BACKGROUND Juvenile amyotrophic lateral sclerosis (JALS) is a rare form of motor neuron disease and occurs before 25 years of age. Only a few cases of juvenile-onset ALS have been reported. MATERIAL AND METHODS To study genetic and clinicopathological features in Chinese patients with juvenile ALS, we retrospectively reviewed ALS patients in our hospital and screened out 2 patients with disease onset before the age of 25. Genetic analysis was carried out with next-generation sequencing (NGS) to identify ALS causative genes. Sanger sequencing was used to validate identified variants. The clinical, electrophysiological, and pathological data were summarized. RESULTS A novel frameshift mutation c.1510dupG (p.G505Wfs*12) was found in Patient One using next-generation sequencing (NGS). Patient Two had a reported pathogenic mutation c.C1483T(p.R495X) with NGS. The mother of Patient Two carried the same mutation as her son and disease onset was at 1.5 years after the death of her son. CONCLUSIONS We identified a novel frameshift mutation associated with JALS. JALS and generally typical ALS, with the same FUS mutation, can appear in a family and present a phenomenon of anticipation. For diagnosis of central nervous system degeneration in adolescents with bulbar symptoms, great attention should be paid to JALS.


Amyotrophic Lateral Sclerosis/genetics , RNA-Binding Protein FUS/genetics , Adolescent , Adult , Asian People/genetics , China , Female , Genetic Testing , High-Throughput Nucleotide Sequencing , Humans , Male , Mutation , RNA-Binding Protein FUS/physiology , Retrospective Studies
11.
Bratisl Lek Listy ; 119(10): 660-663, 2018.
Article En | MEDLINE | ID: mdl-30345776

BACKGROUND: FUS reduces the proliferator factors such as cyclin D1 and Cdk6, and increases Cdk and p27. Therefore, FUS prevents the growth of prostate cancer cells. METHODS: This review tried to summarize data about FUS gene expression in correlation with the degree of prostate cancer. To find the relevant studies, the search in PubMed, Science Direct, and Scopus were performed. RESULTS: Increasing the expression of FUS decreases and increases the rate of apoptosis of prostate cancer cells, respectively. In fact, FUS reduces the proliferator factors such as: cyclin D1 and Cdk6, and increases Cdk (an anti-proliferation factor) and p27 (a proliferative inhibitory factor). Therefore, FUS prevents the growth of prostate cancer cells. An immuno-histochemical analysis showed that FUS gene expression had an inverse correlation with the degree of prostate cancer, which suggests that patients with higher levels of FUS are more likely to survive and less likely to have bone pain. CONCLUSION: The key to FUS is the signaling of the androgen receptor and the progression of the cell cycle in prostate cancer. Based on these findings, we might be able to consider exogenous expression of FUS as a treatment for prostate cancer (Fig. 1, Ref. 32).


Prostatic Neoplasms , RNA-Binding Protein FUS/physiology , Apoptosis , Cell Cycle , Cell Line, Tumor , Humans , Male , Prostatic Neoplasms/genetics , Receptors, Androgen/metabolism
12.
Cell ; 174(3): 688-699.e16, 2018 07 26.
Article En | MEDLINE | ID: mdl-29961577

Proteins such as FUS phase separate to form liquid-like condensates that can harden into less dynamic structures. However, how these properties emerge from the collective interactions of many amino acids remains largely unknown. Here, we use extensive mutagenesis to identify a sequence-encoded molecular grammar underlying the driving forces of phase separation of proteins in the FUS family and test aspects of this grammar in cells. Phase separation is primarily governed by multivalent interactions among tyrosine residues from prion-like domains and arginine residues from RNA-binding domains, which are modulated by negatively charged residues. Glycine residues enhance the fluidity, whereas glutamine and serine residues promote hardening. We develop a model to show that the measured saturation concentrations of phase separation are inversely proportional to the product of the numbers of arginine and tyrosine residues. These results suggest it is possible to predict phase-separation properties based on amino acid sequences.


RNA-Binding Protein FUS/genetics , RNA-Binding Proteins/physiology , Amino Acid Sequence , Amino Acids/chemistry , Animals , Arginine/chemistry , Computer Simulation , HeLa Cells , Humans , Intrinsically Disordered Proteins/genetics , Intrinsically Disordered Proteins/physiology , Phase Transition , Prion Proteins/chemistry , Prion Proteins/genetics , Prions/genetics , Prions/physiology , Protein Domains , RNA-Binding Protein FUS/physiology , RNA-Binding Proteins/isolation & purification , Sf9 Cells , Tyrosine/chemistry
13.
J Hematol Oncol ; 11(1): 77, 2018 06 04.
Article En | MEDLINE | ID: mdl-29866190

BACKGROUND: Despite the overwhelming number of investigations on AKT, little is known about lncRNA on AKT regulation, especially in GBM cells. METHODS: RNA-binding protein immunoprecipitation assay (RIP) and RNA pulldown were used to confirm the binding of LINC00470 and fused in sarcoma (FUS). Confocal imaging, co-immunoprecipitation (Co-IP) and GST pulldown assays were used to detect the interaction between FUS and AKT. EdU assay, CCK-8 assay, and intracranial xenograft assays were performed to demonstrate the effect of LINC00470 on the malignant phenotype of GBM cells. RT-qPCR and Western blotting were performed to test the effect of LINC00470 on AKT and pAKT. RESULTS: In this study, we demonstrated that LINC00470 was a positive regulator for AKT activation in GBM. LINC00470 bound to FUS and AKT to form a ternary complex, anchoring FUS in the cytoplasm to increase AKT activity. Higher pAKT activated by LINC00470 inhibited ubiquitination of HK1, which affected glycolysis, and inhibited cell autophagy. Furthermore, higher LINC00470 expression was associated with GBM tumorigenesis and poor patient prognosis. CONCLUSIONS: Our findings revealed a noncanonical AKT activation signaling pathway, i.e., LINC00470 directly interacts with FUS, serving as an AKT activator to promote GBM progression. LINC00470 has an important referential significance to evaluate the prognosis of patients.


Autophagy , Glioblastoma/pathology , Proto-Oncogene Proteins c-akt/metabolism , RNA, Long Noncoding/physiology , Cell Line, Tumor , Cytoplasm/genetics , Cytoplasm/metabolism , Disease Progression , Humans , Prognosis , Protein Binding , RNA-Binding Protein FUS/metabolism , RNA-Binding Protein FUS/physiology , Signal Transduction
14.
Proc Natl Acad Sci U S A ; 113(48): E7701-E7709, 2016 11 29.
Article En | MEDLINE | ID: mdl-27849576

Amyotrophic lateral sclerosis (ALS) is a progressive motor neuron dysfunction disease that leads to paralysis and death. There is currently no established molecular pathogenesis pathway. Multiple proteins involved in RNA processing are linked to ALS, including FUS and TDP43, and we propose a disease mechanism in which loss of function of at least one of these proteins leads to an accumulation of transcription-associated DNA damage contributing to motor neuron cell death and progressive neurological symptoms. In support of this hypothesis, we find that FUS or TDP43 depletion leads to increased sensitivity to a transcription-arresting agent due to increased DNA damage. Thus, these proteins normally contribute to the prevention or repair of transcription-associated DNA damage. In addition, both FUS and TDP43 colocalize with active RNA polymerase II at sites of DNA damage along with the DNA damage repair protein, BRCA1, and FUS and TDP43 participate in the prevention or repair of R loop-associated DNA damage, a manifestation of aberrant transcription and/or RNA processing. Gaining a better understanding of the role(s) that FUS and TDP43 play in transcription-associated DNA damage could shed light on the mechanisms underlying ALS pathogenesis.


Amyotrophic Lateral Sclerosis/genetics , DNA Breaks, Double-Stranded , DNA-Binding Proteins/physiology , RNA-Binding Protein FUS/physiology , Cell Line , Humans , Motor Neurons/metabolism , Protein Transport , Transcription, Genetic
15.
Adv Exp Med Biol ; 907: 263-96, 2016.
Article En | MEDLINE | ID: mdl-27256390

RNA granules are microscopically visible cellular structures that aggregate by protein-protein and protein-RNA interactions. Using stress granules as an example, we discuss the principles of RNA granule formation, which rely on the multivalency of RNA and multi-domain proteins as well as low-affinity interactions between proteins with prion-like/low-complexity domains (e.g. FUS and TDP-43). We then explore how dysregulation of RNA granule formation is linked to neurodegenerative diseases, such as amyotrophic lateral sclerosis (ALS) and frontotemporal lobar degeneration (FTLD), and discuss possible strategies for therapeutic intervention.


Amyotrophic Lateral Sclerosis/genetics , Cytoplasmic Granules/metabolism , DNA-Binding Proteins/physiology , Frontotemporal Lobar Degeneration/genetics , RNA-Binding Proteins/metabolism , RNA/metabolism , Amyotrophic Lateral Sclerosis/pathology , Animals , Cell Compartmentation , DNA-Binding Proteins/genetics , Frontotemporal Lobar Degeneration/pathology , Humans , Models, Biological , Molecular Chaperones/physiology , Mutation, Missense , Oxidative Stress , Peptide Chain Initiation, Translational , Phase Transition , Protein Binding , Protein Domains , Protein Processing, Post-Translational , RNA-Binding Protein FUS/physiology
16.
J Neurochem ; 138 Suppl 1: 95-111, 2016 08.
Article En | MEDLINE | ID: mdl-27015757

The multiple roles played by RNA binding proteins in neurodegeneration have become apparent following the discovery of TAR DNA binding protein 43 kDa (TDP-43) and fused in sarcoma/translocated in liposarcoma (FUS/TLS) involvement in amyotrophic lateral sclerosis and frontotemporal lobar dementia. In these two diseases, the majority of patients display the presence of aggregated forms of one of these proteins in their brains. The study of their functional properties currently represents a very promising target for developing the effective therapeutic options that are still lacking. This aim, however, must be preceded by an accurate evaluation of TDP-43 and FUS/TLS biological functions, both in physiological and disease conditions. Recent findings have uncovered several aspects of RNA metabolism that can be affected by misregulation of these two proteins. Progress has also been made in starting to understand how the aggregation of these proteins occurs and spreads from cell to cell. The aim of this review will be to provide a general overview of TDP-43 and FUS/TLS proteins and to highlight their physiological functions. At present, the emerging picture is that TDP-43 and FUS/TLS control several aspects of an mRNA's life, but they can also participate in DNA repair processes and in non-coding RNA metabolism. Although their regulatory activities are similar, they regulate mainly distinct RNA targets and show different pathogenetic mechanisms in amyotrophic lateral sclerosis/frontotemporal lobar dementia diseases. The identification of key events in these processes represents today the best chance of finding targetable options for therapeutic approaches that might actually make a difference at the clinical level. The two major RNA Binding Proteins involved in Amyotrophic Lateral Sclerosisi and Frontotemporal Dementia are TDP-43 and FUST/TLS. Both proteins are involved in regulating all aspects of RNA and RNA life cycle within neurons, from transcription, processing, and transport/stability to the formation of cytoplasmic and nuclear stress granules. For this reason, the aberrant aggregation of these factors during disease can impair multiple RNA metabolic pathways and eventually lead to neuronal death/inactivation. The purpose of this review is to provide an up-to-date perspective on what we know about this issue at the molecular level. This article is part of the Frontotemporal Dementia special issue.


DNA-Binding Proteins/genetics , DNA-Binding Proteins/physiology , Frontotemporal Dementia/genetics , RNA-Binding Protein FUS/genetics , RNA-Binding Protein FUS/physiology , TDP-43 Proteinopathies/genetics , TDP-43 Proteinopathies/pathology , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/physiopathology , Frontotemporal Dementia/physiopathology , Frontotemporal Lobar Degeneration/metabolism , Humans
17.
FEBS J ; 283(8): 1475-87, 2016 Apr.
Article En | MEDLINE | ID: mdl-26896672

Translocated in liposarcoma/fused in sarcoma (TLS/FUS) is an RNA-binding protein that regulates the splicing pattern of mRNA transcripts and is known to cause a type of familial amyotrophic lateral sclerosis (ALS). In the absence of TLS, Mammalian enabled (Mena), an actin-regulatory protein and a target of TLS, undergoes preferential alternative splicing. In the present study, we show that the ablation of TLS dysregulates the subcellular location and functions of Mena. When TLS knockout (KO) mouse embryonic fibroblasts (MEFs) were transfected with wild-type Mena, it no longer accumulated at focal adhesions and peripheral structures, whereas the localization of the alternatively spliced form was maintained. Additionally, the ability of Mena to suppress the motility of cells was lost in TLS KO MEFs. Moreover, Mena failed to promote neurite outgrowth in TLS KO primary neurons. Taken together, TLS is intimately involved in the local cytoskeletal dynamics surrounding Mena in both fibroblasts and neurons. The robust change in cytoskeletal dynamics, as indicated by the dysregulation of Mena in TLS KO cells, provides a new insight into the pathogenesis of certain types of ALS.


Actin Cytoskeleton/physiology , Cytoskeletal Proteins/metabolism , Embryo, Mammalian/metabolism , Fibroblasts/metabolism , Neurons/metabolism , RNA-Binding Protein FUS/physiology , Amino Acid Sequence , Animals , Blotting, Western , Cell Differentiation , Cells, Cultured , Cytoskeletal Proteins/genetics , Embryo, Mammalian/cytology , Female , Fibroblasts/cytology , Mice , Mice, Inbred C57BL , Mice, Knockout , Microfilament Proteins , Microscopy, Fluorescence , Molecular Sequence Data , Mutation , Neurons/cytology , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
18.
Oncogene ; 35(15): 1965-76, 2016 Apr 14.
Article En | MEDLINE | ID: mdl-26148230

The ETS transcription factor ERG has been implicated as a major regulator of both normal and aberrant hematopoiesis. In acute myeloid leukemias harboring t(16;21), ERG function is deregulated due to a fusion with FUS/TLS resulting in the expression of a FUS-ERG oncofusion protein. How this oncofusion protein deregulates the normal ERG transcription program is unclear. Here, we show that FUS-ERG acts in the context of a heptad of proteins (ERG, FLI1, GATA2, LYL1, LMO2, RUNX1 and TAL1) central to proper expression of genes involved in maintaining a stem cell hematopoietic phenotype. Moreover, in t(16;21) FUS-ERG co-occupies genomic regions bound by the nuclear receptor heterodimer RXR:RARA inhibiting target gene expression and interfering with hematopoietic differentiation. All-trans retinoic acid treatment of t(16;21) cells as well as FUS-ERG knockdown alleviate the myeloid-differentiation block. Together, the results suggest that FUS-ERG acts as a transcriptional repressor of the retinoic acid signaling pathway.


Chromosomes, Human, Pair 16/genetics , Chromosomes, Human, Pair 21/genetics , Gene Expression Regulation, Neoplastic/genetics , Hematopoiesis/physiology , Leukemia, Myeloid, Acute/genetics , Leukemia, Myelomonocytic, Acute/genetics , Neoplasm Proteins/physiology , Oncogene Proteins, Fusion/physiology , RNA-Binding Protein FUS/physiology , Signal Transduction/physiology , Translocation, Genetic , Tretinoin/physiology , Amino Acid Motifs , Cell Line, Tumor , Chromosomes, Human, Pair 16/ultrastructure , Chromosomes, Human, Pair 21/ultrastructure , Dimerization , Enhancer Elements, Genetic , Hematopoietic Stem Cells/pathology , Humans , Leukemia, Myeloid, Acute/pathology , Leukemia, Myeloid, Acute/physiopathology , Leukemia, Myelomonocytic, Acute/pathology , Leukemia, Myelomonocytic, Acute/physiopathology , Multiprotein Complexes , Neoplasm Proteins/genetics , Neoplastic Stem Cells/pathology , Oncogene Proteins, Fusion/antagonists & inhibitors , Oncogene Proteins, Fusion/genetics , Promoter Regions, Genetic , Protein Binding , Protein Interaction Mapping , Proto-Oncogene Proteins/metabolism , RNA Interference , RNA, Small Interfering/genetics , RNA-Binding Protein FUS/antagonists & inhibitors , RNA-Binding Protein FUS/genetics , Receptors, Retinoic Acid/metabolism , Retinoic Acid Receptor alpha , Retinoid X Receptors/metabolism , Signal Transduction/drug effects , Trans-Activators/metabolism , Transcription Factors/metabolism , Tretinoin/pharmacology , U937 Cells
19.
Cell Death Dis ; 6: e1943, 2015 Oct 22.
Article En | MEDLINE | ID: mdl-26492376

Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disorder due to motor neuron loss. Fused in sarcoma (FUS) protein carrying ALS-associated mutations localizes to stress granules and causes their coalescence into larger aggregates. Here we show that Pur-alpha physically interacts with mutated FUS in an RNA-dependent manner. Pur-alpha colocalizes with FUS carrying mutations in stress granules of motoneuronal cells differentiated from induced pluripotent stem cells and that are derived from ALS patients. We observe that both Pur-alpha and mutated FUS upregulate phosphorylation of the translation initiation factor eukaryotic translation initiation factor 2 alpha and consistently inhibit global protein synthesis. In vivo expression of Pur-alpha in different Drosophila tissues significatively exacerbates the neurodegeneration caused by mutated FUS. Conversely, the downregulation of Pur-alpha in neurons expressing mutated FUS significatively improves fly climbing activity. All these findings suggest that Pur-alpha, through the control of mRNA translation, might be involved in the pathogenesis of ALS associated with the mutation of FUS, and that an alteration of protein synthesis may be directly implicated in the disease. Finally, in vivo RNAi-mediated ablation of Pur-alpha produced locomotion defects in Drosophila, indicating a pivotal role for this protein in the motoneuronal function.


Amyotrophic Lateral Sclerosis/genetics , DNA-Binding Proteins/physiology , Drosophila Proteins/physiology , Nerve Tissue Proteins/physiology , RNA-Binding Protein FUS/physiology , Transcription Factors/physiology , Amyotrophic Lateral Sclerosis/pathology , Animals , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Drosophila/genetics , Drosophila/physiology , Drosophila Proteins/antagonists & inhibitors , Drosophila Proteins/genetics , Eukaryotic Initiation Factor-2/metabolism , HeLa Cells , Humans , Induced Pluripotent Stem Cells , Motor Neurons/metabolism , Mutation , Nerve Tissue Proteins/metabolism , Phosphorylation , Protein Biosynthesis/genetics , RNA Interference , RNA, Messenger/metabolism , RNA-Binding Protein FUS/genetics , RNA-Binding Protein FUS/metabolism , Ribosomes/metabolism , Transcription Factors/antagonists & inhibitors , Transcription Factors/genetics , Transcription Factors/metabolism
20.
Biochim Biophys Acta ; 1849(12): 1398-410, 2015 Dec.
Article En | MEDLINE | ID: mdl-26514432

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.


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
...