ArticlePublisher preview available

Monomethylated and unmethylated FUS exhibit increased binding to Transportin and distinguish FTLD-FUS from ALS-FUS

Authors:
  • BarcelonaBeta Brain Research Center (BBRC)
To read the full-text of this research, you can request a copy directly from the authors.

Abstract and Figures

Deposition of the nuclear DNA/RNA-binding protein Fused in sarcoma (FUS) in cytosolic inclusions is a common hallmark of some cases of frontotemporal lobar degeneration (FTLD-FUS) and amyotrophic lateral sclerosis (ALS-FUS). Whether both diseases also share common pathological mechanisms is currently unclear. Based on our previous finding that FUS deposits are hypomethylated in FTLD-FUS but not in ALS-FUS, we have now investigated whether genetic or pharmacological inactivation of Protein arginine methyltransferase 1 (PRMT1) activity results in unmethylated FUS or in alternatively methylated forms of FUS. To do so, we generated FUS-specific monoclonal antibodies that specifically recognize unmethylated arginine (UMA), monomethylated arginine (MMA) or asymmetrically dimethylated arginine (ADMA). Loss of PRMT1 indeed not only results in an increase of UMA FUS and a decrease of ADMA FUS, but also in a significant increase of MMA FUS. Compared to ADMA FUS, UMA and MMA FUS exhibit much higher binding affinities to Transportin-1, the nuclear import receptor of FUS, as measured by pull-down assays and isothermal titration calorimetry. Moreover, we show that MMA FUS occurs exclusively in FTLD-FUS, but not in ALS-FUS. Our findings therefore provide additional evidence that FTLD-FUS and ALS-FUS are caused by distinct disease mechanisms although both share FUS deposits as a common denominator.
Characterization of monoclonal antibodies specific for UMA and MMA FUS. a Schematic figure of the FUS protein. FUS contains multiple functional domains: a QGSY-rich transcriptional activation domain (glutamine, glycine, serine and tyrosine rich domain), three RGG domains (arginine-glycine-glycine repeats); a RRM domain (RNA recognition motif), a ZnF domain (zinc finger domain), and the PY-NLS (proline-tyrosine nuclear localization signal). We generated antibodies against the differently methylated RGG3 domain (FUS473–503) with all nine arginines in three different forms of methylation modifications: UMA (red), MMA (blue) or ADMA (green). An asterisk indicates a methyl group. b Schematic representation of arginine methylation steps. All PRMTs (type I, II and III) are capable to add a methyl group and generate a monomethylated arginine (MMA). Subsequently, type I PRMTs deposit the asymmetrically dimethylated arginine (ADMA) signature, whereas type II PRMTs deposit the symmetrically dimethylated ariginine (SMDA) signature. Type III PRMT (PRMT7) can only generate MMA. c HeLa cells were transfected with siRNAs against FUS, EWS, TAF-15 or a non-targeting control and were subsequently treated with AdOx or left untreated. 48 h post-transfection cell lysates were analysed by immunoblotting with the antibodies raised against UMA FUS (14G1), MMA FUS (15E11) and ADMA (9G6). All of them show a signal at the predicted molecular weight of FUS (arrow) that disappears upon knockdown of FUS. Whereas ADMA FUS antibody (9G6) shows immunoreactivity to untreated cells, UMA FUS (14G1) and MMA FUS (15E11) antibodies only reacted with lysates derived from AdOx-treated cells. These antibodies do not show cross-reactivity against EWS and TAF-15. Tubulin was used as a loading control and FUS, EWS and TAF-15 antibodies were used to examine knockdown efficiency. Asterisks: unspecific bands
… 
Loss of PRMT1 elicits a switch in the methylation pattern of FUS from ADMA to MMA and UMA. a Whole cell lysates of PRMT1 knockout (PRMT1 −/−) and wild-type (PRMT1 +/+) mES cells (duplicates from two independent experiments) were prepared and immunoblotted with antibodies specific for ADMA FUS (9G6), MMA FUS (15E11) and UMA FUS (14G1). A decrease in ADMA FUS paralleled by an increase in UMA and MMA FUS is observed in PRMT1 −/− mES cells. Membranes were blotted with anti-Tubulin and anti-total FUS antibodies to visualize equal loading and with a PRMT1-specific antibody to confirm PRMT1 knockout. Asterisks indicate unspecific bands. b, c Whole cell lysates were subjected to immunoprecipitation (IP) with an anti-FUS antibody and an isotype control antibody. Precipitated proteins were analysed by immunoblotting (IB) with a general anti-MMA antibody (D5A12) that recognizes monomethylated RGG motifs. This demonstrates an increase in MMA FUS in PRMT1 −/− cells (top panel). The input (whole cell lysates) is shown in the left panel. Equal amounts of FUS protein were immunoprecipitated from PRMT1 +/+ and PRMT1 −/− cells (bottom panel). The same immunoprecipitated material was also immunoblotted with another anti-MMA antibody (Me-R4-100) raised against monomethylated arginine. This confirmed an increase in MMA FUS upon PRMT1 knockout (PRMT1 −/−). d Immunoprecipitation (IP) of monomethylated proteins followed by immunoblotting (IB) for FUS confirmed an increase of MMA FUS in PRMT1 −/− cells
… 
This content is subject to copyright. Terms and conditions apply.
1 3
Acta Neuropathol (2016) 131:587–604
DOI 10.1007/s00401-016-1544-2
ORIGINAL PAPER
Monomethylated and unmethylated FUS exhibit increased
binding to Transportin and distinguish FTLD‑FUS
from ALS‑FUS
Marc Suárez‑Calvet1,7,9 · Manuela Neumann3,4 · Thomas Arzberger5,6,7 ·
Claudia Abou‑Ajram1,2 · Eva Funk1 · Hannelore Hartmann7,8 · Dieter Edbauer7,8 ·
Elisabeth Kremmer10 · Christoph Göbl11,12 · Moritz Resch11,12 ·
Benjamin Bourgeois11,12 · Tobias Madl11,12,13,14 · Stefan Reber15,16 ·
Daniel Jutzi15 · Marc‑David Ruepp15 · Ian R. A. Mackenzie17 · Olaf Ansorge18 ·
Dorothee Dormann1,2,8 · Christian Haass1,7,8
Received: 31 July 2015 / Revised: 29 January 2016 / Accepted: 29 January 2016 / Published online: 19 February 2016
© Springer-Verlag Berlin Heidelberg 2016
previous finding that FUS deposits are hypomethylated in
FTLD-FUS but not in ALS-FUS, we have now investigated
whether genetic or pharmacological inactivation of Pro-
tein arginine methyltransferase 1 (PRMT1) activity results
in unmethylated FUS or in alternatively methylated forms
of FUS. To do so, we generated FUS-specific monoclonal
antibodies that specifically recognize unmethylated argi-
nine (UMA), monomethylated arginine (MMA) or asym-
metrically dimethylated arginine (ADMA). Loss of PRMT1
indeed not only results in an increase of UMA FUS and a
Abstract Deposition of the nuclear DNA/RNA-binding
protein Fused in sarcoma (FUS) in cytosolic inclusions is
a common hallmark of some cases of frontotemporal lobar
degeneration (FTLD-FUS) and amyotrophic lateral sclero-
sis (ALS-FUS). Whether both diseases also share common
pathological mechanisms is currently unclear. Based on our
Electronic supplementary material The online version of this
article (doi:10.1007/s00401-016-1544-2) contains supplementary
material, which is available to authorized users.
* Dorothee Dormann
dorothee.dormann@med.uni-muenchen.de
* Christian Haass
christian.haass@mail03.med.uni-muenchen.de
1 Biomedical Center (BMC), Biochemistry, Ludwig-
Maximilians-University Munich, Feodor-Lynen Strasse 17
81377 Munich, Germany
2 Present Address: BioMedical Center (BMC), Lehrstuhl
Zellbiologie (Anatomie III), Großhaderner Strasse 9,
82152 Planegg-Martinsried, Germany
3 Department of Neuropathology, University of Tübingen,
72076 Tübingen, Germany
4 DZNE, German Center for Neurodegenerative Diseases,
72076 Tübingen, Germany
5 Department of Psychiatry and Psychotherapy, Ludwig-
Maximilians-University Munich, 80336 Munich, Germany
6 Center for Neuropathology and Prion Research, Ludwig-
Maximilians-University Munich, 81377 Munich, Germany
7 German Center for Neurodegenerative Diseases (DZNE)
Munich, Feodor-Lynen Strasse 17, 81377 Munich, Germany
8 Munich Cluster for Systems Neurology (SyNergy),
81377 Munich, Germany
9 Universitat Autònoma de Barcelona, 08193 Bellaterra,
Barcelona, Spain
10 Institute of Molecular Immunology, Helmholtz Zentrum
München, German Research Center for Environmental
Health (GmbH), 81377 Munich, Germany
11 Department of Chemistry, Center for Integrated Protein
Science Munich (CIPSM), Technische Universität München,
Lichtenbergstr.4, 85747 Garching, Germany
12 Institute of Structural Biology, Helmholtz Zentrum München,
85764 Neuherberg, Germany
13 Institute of Molecular Biology and Biochemistry, Center
of Molecular Medicine, Medical University of Graz,
8010 Graz, Austria
14 Omics Center Graz, BioTechMed, 8010 Graz, Austria
15 Department of Chemistry and Biochemistry, University
of Bern, 3012 Bern, Switzerland
16 Graduate School for Cellular and Biomedical Sciences,
University of Bern, 3012 Bern, Switzerland
17 Department of Pathology, Vancouver General Hospital,
University of British Columbia, Vancouver, Canada
18 Department of Neuropathology, John Radcliffe Hospital,
Oxford, UK
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
... Arginine methylation is an important post-translational modification (PTM) that provides a major level of epigenetic regulation via histone methylation and is found on many non-histone RNA-binding proteins (RBPs) regulating RNA processing and splicing (14)(15)(16)(17)(18). Recently, arginine methylation of several RBPs has been linked to neurodegenerative disorders, such as spinal and bulbar muscular atrophy (SBMA), amyotrophic lateral sclerosis (ALS) and frontotemporal lobar degeneration (FTLD) (19)(20)(21). For example, methylation of RBP FUS (fused in sarcoma) modulates its cytoplasmic mislocalization in ALS (22) and FTLD (23). RNA processing and splicing abnormalities have been also associated with HD based on several expression profiling studies in HD models and recent proteomics screens (10,(24)(25)(26)(27)(28). ...
... These experiments suggest that, as previously reported (50,(56)(57)(58), TAF15, HNRNPUL1 and SAM68 are substrates for PRMT1, as evidenced by an increase in arginine methylation signal upon PRMT1 overexpression. We were not able to detect an increase in arginine methylated FUS with any PRMT tested under these conditions, although FUS has been reported to be a PRMT1 substrate as well (23,59). Notably, we found that TAF15 may be also methylated by PRMT6, which interacts and co-localizes with HTT in the nucleus (30). ...
... Although arginine methylation has been recently linked to neurodegeneration (17,(19)(20)(21)(22)(23), this is the first study conducted using neuronal models and addressing proteome-wide changes in arginine methylation in a neurodegenerative disease. An important conclusion from both our proteomic and metabolomic analyses is that protein arginine methylation is significantly altered in a patient-derived neuronal HD model with most detected arginine methylated proteins hypomethylated in HD relative to normal control. ...
Article
Full-text available
Huntington's disease (HD) is a progressive neurodegenerative disorder caused by a CAG repeat expansion in the HD gene, coding for huntingtin protein (HTT). Mechanisms of HD cellular pathogenesis remain undefined and likely involve disruptions in many cellular processes and functions presumably mediated by abnormal protein interactions of mutant HTT. We previously found HTT interaction with several protein arginine methyl-transferases (PRMT) enzymes. Protein arginine methylation mediated by PRMT enzymes is an important post-translational modification with an emerging role in neurodegeneration. We found that normal (but not mutant) HTT can facilitate the activity of PRMTs in vitro and the formation of arginine methylation complexes. These interactions appear to be disrupted in HD neurons. This suggests an additional functional role for HTT/PRMT interactions, not limited to substrate/enzyme relationship, which may result in global changes in arginine protein methylation in HD. Our quantitative analysis of striatal precursor neuron proteome indicated that arginine protein methylation is significantly altered in HD. We identified a cluster highly enriched in RNA-binding proteins with reduced arginine methylation, which is essential to their function in RNA processing and splicing. We found that several of these proteins interact with HTT, and their RNA-binding and localization is affected in HD cells likely due to a compromised arginine methylation and/or abnormal interactions with mutant HTT. These studies reveal a potential new mechanism for disruption of RNA processing in HD, involving a direct interaction of HTT with methyl-transferase enzymes and modulation of their activity, and highlighting methylation of arginine as potential new therapeutic target for HD.
... Nevertheless, FUS-ADMA has been observed to alter FUS nucleocytoplasmic localization, leading to accumulated protein in the cytoplasm due to decreased binding affinity with TnPO1/Kβ2 as a result of its methylation interfering with its protein-protein binding properties (Dormann et al., 2012;Tradewell et al., 2012;Jäckel et al., 2015;Suarez-Calvet et al., 2016). Interestingly, Suárez-Calvet and collaborators observed differential FUS methylation in ALS-FUS and FTD-FUS inclusions from brain samples, where ALS-FUS inclusions contained exclusively FUS-ADMA forms, whereas FTD-FUS inclusions contained mono-or unmethylated forms of FUS (Suarez-Calvet et al., 2016), suggesting differential pathogenic mechanisms for these two diseases. ...
... Nevertheless, FUS-ADMA has been observed to alter FUS nucleocytoplasmic localization, leading to accumulated protein in the cytoplasm due to decreased binding affinity with TnPO1/Kβ2 as a result of its methylation interfering with its protein-protein binding properties (Dormann et al., 2012;Tradewell et al., 2012;Jäckel et al., 2015;Suarez-Calvet et al., 2016). Interestingly, Suárez-Calvet and collaborators observed differential FUS methylation in ALS-FUS and FTD-FUS inclusions from brain samples, where ALS-FUS inclusions contained exclusively FUS-ADMA forms, whereas FTD-FUS inclusions contained mono-or unmethylated forms of FUS (Suarez-Calvet et al., 2016), suggesting differential pathogenic mechanisms for these two diseases. ...
... Mechanistically TnPO blocks FUS phase transition by binding to the C-terminal RGG3-PY domain arginines, interfering with their phase transition inducing properties. Finally, they observed that arginines methylation of the C-terminal RGG3-PY domain reduced FUS LLPS and accumulation in SGs (Hofweber et al., 2018), which correlates with the observations in FTD-FUS patients' inclusion where FUS is unmethylated (Dormann et al., 2012;Suarez-Calvet et al., 2016). ...
Thesis
Amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD) are two untreatable neurodegenerative diseases. Even thought they are two distinctive diseases, they share a series of clinical, genetic and histological hallmarks, thus defining the ALS-FTD continuum. Mutations in the FUS gene have been linked with ALS, whereas alterations in FUS proteins have been detected in FTD patients. Both diseases are characterized by the presence of cytosolic FUS aggregates.We have studied the autoregulation mechanisms of FUS in a mouse model via de activation of an alternative splicing pathway by the insertion of a human wild-type FUS transgene, which has allowed us to potentially elucidate new therapeutic approaches by gene therapy. Furthermore, our mice develop FTD-like symptoms. Our results suggest an alteration in cortical synapses which could originate the observed cognitive and behavioural deficits, accompanied by alterations in the cholinergic system.
... In fALS-FUS the increased propensity of FUS to form irreversible condensates is largely driven by the presence of missense mutations. In contrast, in FTLD-FUS the pathological condensation is driven by hypomethylation of arginine residues in FUS [8,9]. These differences in how the pathological condensates are formed raises the possibility that there may also be distinctions in the molecular mechanism(s) by which they induce neuronal dysfunction. ...
... To date, much of the research exploring the mechanism of FUS pathology in neurodegeneration has focused on the missense mutations associated with fALS-FUS [10]. Much less is currently known about the molecular/ cellular pathobiology of hypomethylated FUS associated with sporadic FTLD-FUS [9]. ...
Article
Full-text available
The hypomethylation of fused in sarcoma (FUS) in frontotemporal lobar degeneration promotes the formation of irreversible condensates of FUS. However, the mechanisms by which these hypomethylated FUS condensates cause neuronal dysfunction are unknown. Here we report that expression of FUS constructs mimicking hypomethylated FUS causes aberrant dendritic FUS condensates in CA1 neurons. These hypomethylated FUS condensates exhibit spontaneous, and activity induced movement within the dendrite. They impair excitatory synaptic transmission, postsynaptic density-95 expression, and dendritic spine plasticity. These neurophysiological defects are dependent upon both the dendritic localisation of the condensates, and their ability to undergo liquid–liquid phase separation. These results indicate that the irreversible liquid–liquid phase separation is a key component of hypomethylated FUS pathophysiology in sporadic FTLD, and this can cause synapse dysfunction in sporadic FTLD.
... Many recent studies have concentrated on the PTM of FUS-pathology, and we believe it is linked to the ALS-FUS pathology mechanism. PTM has been identified as an important protein regulator in neurodegenerative diseases such as Alzheimer's disease, Amyotrophic lateral sclerosis, Parkinson's disease, and Huntington's disease (Schaffert and Carter 2020;Suárez-Calvet et al. 2016;Sternburg et al. 2022). ...
... Page 7 of 10 Jun et al. Journal of Analytical Science and Technology (2023) 14:23 Studies on FUS PTM have revealed hypomethylation of FUS in FTD-FUS patients unrelated to FUS mutations and unmethylation of FUS in ALS-FUS patients (Suárez-Calvet et al. 2016). The majority of these studies have focused on FUS methylation, but it is still unknown why FUS aggregates in the cytoplasm rather than the nucleus. ...
Article
Full-text available
Mutations in the fused-in-sarcoma (FUS) gene have been linked to familial amyotrophic lateral sclerosis (fALS). FUS aggregates in the cytosol and associates with stress granules (SGs) in pathological cases, whereas FUS is normally found in the nucleus. However, little is known about how FUS mutations cause neurodegeneration in ALS, which is distinguished by FUS-positive inclusion and stress granules. In this study, we investigated the mechanism of abnormal cytoplasmic aggregate formation caused by ALS-linked FUS mutations. FUS R521C interacted more with ubiquitin-associated protein 2-like (UBAP2L) and protein arginine methyltransferase 1 (PRMT1) than FUS WT, and PRMT1 and UBAP2L are sequestered into FUS R521C-positive stress granules under oxidative stress. PRMT1 asymmetrically demethylates UBAP2L, which is required for both SG assembly and disassembly. Furthermore, in FUS R521C, arginine methylation of UBAP2L is reduced, and the loss of PRMT1 increases FUS-positive SGs in oxidative stress. These results imply that an aberrant interaction between FUS-R521C and PRMT1 causes insufficient arginine methylation of UBAP2L, resulting in abnormal FUS-R521C-positive SGs remaining in the cytoplasm. This study could lead to the identification of a new pathogenic mechanism and therapeutic targets for the FUS mutation, which has been associated with abnormally increased protein interactions in ALS. Graphical abstract
Article
Full-text available
Amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD) are fatal neurodegenerative disorders on a disease spectrum that are characterized by the cytoplasmic mislocalization and aberrant phase transitions of prion-like RNA-binding proteins (RBPs). The common accumulation of TAR DNA-binding protein-43 (TDP-43), fused in sarcoma (FUS), and other nuclear RBPs in detergent-insoluble aggregates in the cytoplasm of degenerating neurons in ALS/FTD is connected to nuclear pore dysfunction and other defects in the nucleocytoplasmic transport machinery. Recent advances suggest that beyond their canonical role in the nuclear import of protein cargoes, nuclear-import receptors (NIRs) can prevent and reverse aberrant phase transitions of TDP-43, FUS, and related prion-like RBPs and restore their nuclear localization and function. Here, we showcase the NIR family and how they recognize cargo, drive nuclear import, and chaperone prion-like RBPs linked to ALS/FTD. We also discuss the promise of enhancing NIR levels and developing potentiated NIR variants as therapeutic strategies for ALS/FTD and related neurodegenerative proteinopathies. Graphical Abstract
Article
Our understanding of stress granule (SG) biology has deepened considerably in recent years, and with this increased understanding links have been made between SGs and numerous neurodegenerative diseases. One of the proposed mechanisms by which SGs and any associated protein aggregates may become pathological is based upon defects in their autophagic clearance, and so the precise processes governing the degradation of SGs are important to understand. Mutations and disease‐associated variants implicated in amyotrophic lateral sclerosis, Huntington's disease, Parkinson's disease, and frontotemporal lobar dementia compromise autophagy, whilst autophagy‐inhibiting drugs or knockdown of essential autophagy proteins result in the persistence of SGs. In this review, we will consider the current knowledge regarding the autophagy of stress granules.
Article
Full-text available
Biomolecular condensation, driven by multivalency, serves as a fundamental mechanism within cells, facilitating the formation of distinct compartments, including membraneless organelles that play essential roles in various cellular processes. Perturbations in the delicate equilibrium of condensation, whether resulting in gain or loss of phase separation, have robustly been associated with cellular dysfunction and physiological disorders. As ongoing research endeavors wholeheartedly embrace this newly acknowledged principle, a transformative shift is occurring in our comprehension of disease. Consequently, significant strides have been made in unraveling the profound relevance and potential causal connections between abnormal phase separation and various diseases. This comprehensive review presents compelling recent evidence that highlight the intricate associations between aberrant phase separation and neurodegenerative diseases, cancers, and infectious diseases. Additionally, we provide a succinct summary of current efforts and propose innovative solutions for the development of potential therapeutics to combat the pathological consequences attributed to aberrant phase separation.
Article
Spinal muscular atrophy (SMA) is a leading genetic cause of infant mortality. The advent of approved treatments for this devastating condition has significantly changed SMA patients' life expectancy and quality of life. Nevertheless, these are not without limitations, and research efforts are underway to develop new approaches for improved and long‐lasting benefits for patients. Protein arginine methyltransferases (PRMTs) are emerging as druggable epigenetic targets, with several small‐molecule PRMT inhibitors already in clinical trials. From a screen of epigenetic molecules, we have identified MS023, a potent and selective type I PRMT inhibitor able to promote SMN2 exon 7 inclusion in preclinical SMA models. Treatment of SMA mice with MS023 results in amelioration of the disease phenotype, with strong synergistic amplification of the positive effect when delivered in combination with the antisense oligonucleotide nusinersen. Moreover, transcriptomic analysis revealed that MS023 treatment has minimal off‐target effects, and the added benefit is mainly due to targeting neuroinflammation. Our study warrants further clinical investigation of PRMT inhibition both as a stand‐alone and add‐on therapy for SMA.
Article
Full-text available
Nuclear to cytoplasmic mislocalization and aggregation of multiple RNA-binding proteins (RBPs), including FUS, are the main neuropathological features of the majority of cases of amyotrophic lateral sclerosis (ALS) and frontotemporal lobular degeneration (FTLD). In ALS-FUS, these aggregates arise from disease-associated mutations in FUS, whereas in FTLD-FUS, the cytoplasmic inclusions do not contain mutant FUS, suggesting different molecular mechanisms of FUS pathogenesis in FTLD that remain to be investigated. We have previously shown that phosphorylation of the C-terminal Tyr526 of FUS results in increased cytoplasmic retention of FUS due to impaired binding to the nuclear import receptor TNPO1. Inspired by the above notions, in the current study we developed a novel antibody against the C-terminally phosphorylated Tyr526 FUS (FUSp-Y526) that is specifically capable of recognizing phosphorylated cytoplasmic FUS, which is poorly recognized by other commercially available FUS antibodies. Using this FUSp-Y526 antibody, we demonstrated a FUS phosphorylation-specific effect on the cytoplasmic distribution of soluble and insoluble FUSp-Y526 in various cells and confirmed the involvement of the Src kinase family in Tyr526 FUS phosphorylation. In addition, we found that FUSp-Y526 expression pattern correlates with active pSrc/pAbl kinases in specific brain regions of mice, indicating preferential involvement of cAbl in the cytoplasmic mislocalization of FUSp-Y526 in cortical neurons. Finally, the pattern of immunoreactivity of active cAbl kinase and FUSp-Y526 revealed altered cytoplasmic distribution of FUSp-Y526 in cortical neurons of post-mortem frontal cortex tissue from FTLD patients compared with controls. The overlap of FUSp-Y526 and FUS signals was found preferentially in small diffuse inclusions and was absent in mature aggregates, suggesting possible involvement of FUSp-Y526 in the formation of early toxic FUS aggregates in the cytoplasm that are largely undetected by commercially available FUS antibodies. Given the overlapping patterns of cAbl activity and FUSp-Y526 distribution in cortical neurons, and cAbl induced sequestration of FUSp-Y526 into G3BP1 positive granules in stressed cells, we propose that cAbl kinase is actively involved in mediating cytoplasmic mislocalization and promoting toxic aggregation of wild-type FUS in the brains of FTLD patients, as a novel putative underlying mechanism of FTLD-FUS pathophysiology and progression.
Article
Full-text available
Aims: Transportin 1 (TNPO 1) is an abundant component of the Fused in Sarcoma (FUS)-immunopositive inclusions seen in a subgroup of frontotemporal lobar degeneration (FTLD-FUS). TNPO 1 has been shown to bind to the C-terminal nuclear localizing signal (NLS) of FUS and mediate its nuclear import. Amyotrophic lateral sclerosis (ALS)-linked C-terminal mutants disrupt TNPO 1 binding to the NLS and impair nuclear import in cell culture. If this held true for human ALS then we predicted that FUS inclusions in patients with C-terminal FUS mutations would not colocalize with TNPO 1. Methods: Expression of TNPO 1 and colocalization with FUS was studied in the frontal cortex of FTLD-FUS (n = 3) and brain and spinal cord of ALS-FUS (n = 3), ALS-C9orf72 (n = 3), sporadic ALS (n = 7) and controls (n = 7). Expression levels and detergent solubility of TNPO 1 was measured by Western blot. Results: Aggregates of TNPO 1 were abundant and colocalized with FUS inclusions in the cortex of all FTLD-FUS cases. In contrast, no TNPO 1-positive aggregates or FUS colocalization was evident in two-thirds, ALS-FUS cases and was rare in one ALS-FUS case. Nor were they present in C9orf72 or sporadic ALS. No increase in the levels of TNPO 1 was seen in Western blots of spinal cord tissues from all ALS cases compared with controls. Conclusions: These findings confirm that C-terminal FUS mutations prevent TNPO 1 binding to the NLS, inhibiting nuclear import and promoting cytoplasmic aggregation. The presence of TNPO 1 in wild-type FUS aggregates in FTLD-FUS distinguishes the two pathologies and implicates different disease mechanisms.
Article
Full-text available
Abnormal cytoplasmic accumulation of Fused in Sarcoma (FUS) in neurons defines subtypes of amyotrophic lateral sclerosis (ALS) and frontotemporal lobar degeneration (FTLD). FUS is a member of the FET protein family that includes Ewing's sarcoma (EWS) and TATA-binding protein-associated factor 2N (TAF15). FET proteins are predominantly localized to the nucleus, where they bind RNA and DNA to modulate transcription, mRNA splicing, and DNA repair. In ALS cases with FUS inclusions (ALS-FUS), mutations in the FUS gene cause disease, whereas FTLD cases with FUS inclusions (FTLD-FUS) do not harbor FUS mutations. Notably, in FTLD-FUS, all FET proteins accumulate with their nuclear import receptor Transportin 1 (TRN1), in contrast ALS-FUS inclusions are exclusively positive for FUS. In the present study, we show that induction of DNA damage replicates several pathologic hallmarks of FTLD-FUS in immortalized human cells and primary human neurons and astrocytes. Treatment with the antibiotic calicheamicin γ1, which causes DNA double-strand breaks, leads to the cytoplasmic accumulation of FUS, TAF15, EWS, and TRN1. Moreover, cytoplasmic translocation of FUS is mediated by phosphorylation of its N terminus by the DNA-dependent protein kinase. Finally, we observed elevated levels of phospho-H2AX in FTLD-FUS brains, indicating that DNA damage occurs in patients. Together, our data reveal a novel regulatory mechanism for FUS localization in cells and suggest that DNA damage may contribute to the accumulation of FET proteins observed in human FTLD-FUS cases, but not in ALS-FUS.
Article
Full-text available
The covalent attachment of methyl groups to the side-chain of arginine residues is known to play essential roles in regulation of transcription, protein function and RNA metabolism. The specific N-methylation of arginine residues is catalyzed by a small family of gene products known as protein arginine methyltransferases; however, very little is known about which arginine residues become methylated on target substrates. Here we describe a proteomics methodology that combines single-step immunoenrichment of methylated peptides with high-resolution mass spectrometry to identify endogenous arginine mono-methylation (MMA) sites. We thereby identify 1,027 site-specific MMA sites on 494 human proteins, discovering numerous novel mono-methylation targets and confirming the majority of currently known MMA substrates. Nuclear RNA-binding proteins involved in RNA processing, RNA localization, transcription, and chromatin remodeling are predominantly found modified with MMA. Despite this, MMA sites prominently are located outside RNA-binding domains as compared to the proteome-wide distribution of arginine residues. Quantification of arginine methylation in cells treated with Actinomycin D uncovers strong site-specific regulation of MMA sites during transcriptional arrest. Interestingly, several MMA sites are down-regulated after a few hours of transcriptional arrest. In contrast, the corresponding di-methylation or protein expression level is not altered in expression, confirming that MMA sites contain regulated functions on their own. Collectively, we present a site-specific MMA dataset in human cells and demonstrate for the first time that MMA is a dynamic post-translational modification regulated during transcriptional arrest by a hitherto uncharacterized arginine demethylase.
Article
Full-text available
Protein methylation is a common posttranslational modification that mostly occurs on arginine and lysine residues. Arginine methylation has been reported to regulate RNA processing, gene transcription, DNA damage repair, protein translocation, and signal transduction. Lysine methylation is best known to regulate histone function and is involved in epigenetic regulation of gene transcription. To better study protein methylation, we have developed highly specific antibodies against monomethyl arginine, asymmetric dimethyl arginine, and monomethyl, dimethyl, and trimethyl lysine motifs respectively. These antibodies were used to perform immunoaffinity purification (IAP) of methyl peptides followed by LC-MS/MS analysis to identify and quantify arginine and lysine methylation sites in several model studies. Overall, we identified over 1000 arginine methylation sites in human cell lines and mouse tissues, and approximately 160 lysine methylation sites in human cell line HCT116. The methylation sites that were identified in this study exceed those found in the literature to date. Detailed analysis of arginine-methylated proteins observed in mouse brain compared to those found in mouse embryo shows tissue-specific distribution of arginine methylation, and extends the type of proteins that are known to be arginine methylated to many new protein types. Many arginine-methylated proteins that we identified from the brain including receptors, ion channels, transporters, and vesicle proteins, are involved in synaptic transmission, while the most abundant methylated proteins identified from mouse embryo are transcriptional regulators and RNA processing proteins.
Article
Full-text available
Motifs rich in arginines and glycines were recognized several decades ago to play functional roles and were termed glycine-arginine-rich (GAR) domains and/or RGG boxes. We review here the evolving functions of the RGG box along with several sequence variations that we collectively term the RGG/RG motif. Greater than 1,000 human proteins harbor the RGG/RG motif, and these proteins influence numerous physiological processes such as transcription, pre-mRNA splicing, DNA damage signaling, mRNA translation, and the regulation of apoptosis. In particular, we discuss the role of the RGG/RG motif in mediating nucleic acid and protein interactions, a function that is often regulated by arginine methylation and partner-binding proteins. The physiological relevance of the RGG/RG motif is highlighted by its association with several diseases including neurological and neuromuscular diseases and cancer. Herein, we discuss the evidence for the emerging diverse functionality of this important motif.
Article
Full-text available
Mutations in the gene encoding Fused in Sarcoma (FUS) cause amyotrophic lateral sclerosis (ALS), a fatal neurodegenerative disorder. FUS is a predominantly nuclear DNA and RNA binding protein that is involved in RNA processing. Large FUS immuno-reactive inclusions fill the perikaryon of surviving motor neurons of ALS patients carrying mutations at post-mortem. This sequestration of FUS is predicted to disrupt RNA processing and initiate neurodegeneration. Here we demonstrate that C-terminal ALS mutations disrupt the nuclear localising signal (NLS) of FUS resulting in cytoplasmic accumulation in transfected cells and patient fibroblasts. FUS mislocalisation is rescued by the addition of the wild-type FUS NLS to mutant proteins. We also show that oxidative stress recruits mutant FUS to cytoplasmic stress granules where it is able to bind and sequester wild-type FUS. While FUS interacts with itself directly by protein-protein interaction, the recruitment of FUS to stress granules and interaction with PABP is RNA dependent. These findings support a two-hit hypothesis whereby cytoplasmic mislocalisation of FUS protein, followed by cellular stress contributes to the formation of cytoplasmic aggregates that may sequester FUS, disrupt RNA processing and initiate motor neuron degeneration.
Article
Full-text available
Arginine methylation is a common posttranslational modification that is found on both histone and non-histone proteins. Three types of arginine methylation exist in mammalian cells: monomethylarginine (MMA), asymmetric dimethylarginine (ADMA) and symmetric dimethylarginine (SDMA). PRMT1 is the primary methyltransferase that deposits the ADMA mark, and it accounts for over 90% of this type of methylation. Here, we show that with the loss of PRMT1 activity, there are major increases in global MMA and SDMA levels, as detected by type-specific antibodies. Amino acid analysis confirms that MMA and SDMA levels accumulate when ADMA levels are reduced. These findings reveal the dynamic interplay between different arginine methylation types in the cells, and that the pre-existence of the dominant ADMA mark can block the occurrence of SDMA and MMA marks on the same substrate. This study provides clear evidence of competition for different arginine methylation types on the same substrates.
Article
The nuclear protein fused in sarcoma (FUS) is found in cytoplasmic inclusions in a subset of patients with the neurodegenerative disorder frontotemporal lobar degeneration (FTLD-FUS). FUS contains a methylated arginine-glycine-glycine domain that is required for transport into the nucleus. Recent findings have shown that this domain is hypomethylated in patients with FTLD-FUS. To determine whether the cause of hypomethylation is the result of mutations in protein N-arginine methyltransferases (PRMTs), we selected 3 candidate genes (PRMT1, PRMT3, and PRMT8) and performed complete sequencing analysis and real-time polymerase chain reaction mRNA expression analysis in 20 FTLD-FUS cases. No mutations or statistically significant changes in expression were observed in our patient samples, suggesting that defects in PRMTs are not the cause of FTLD-FUS.