Skip to main content
Log in

Treatment with a Global Methyltransferase Inhibitor Induces the Intranuclear Aggregation of ALS-Linked FUS Mutant In Vitro

  • Original Paper
  • Published:
Neurochemical Research Aims and scope Submit manuscript

Abstract

FUS/TLS (fused in sarcoma/translocated in liposarcoma) encodes a multifunctional DNA/RNA binding protein with non-classical carboxy (C)-terminal nuclear localization signal (NLS). A variety of ALS-linked mutations are clustered in the C-terminal NLS, resulting in the cytoplasmic mislocalization and aggregation. Since the arginine methylations are implicated in the nuclear-cytoplasmic shuttling of FUS, a methylation inhibitor could be one of therapeutic targets for FUS-linked ALS. We here examined effects of methylation inhibitors on the cytoplasmic mislocalization and aggregates of ALS-linked C-terminal FUS mutant in a cell culture system. Treatment with adenosine dialdehyde (AdOx), a representative global methyltransferase inhibitor, remarkably mitigated the cytoplasmic mislocalization and aggregation of FUS mutant, which is consistent with previous reports. However, AdOx treatment of higher concentration and longer time period evoked the intranuclear aggregation of the ectopic expressed FUS protein. The pull down assay and the morphological analysis indicated the binding between FUS and Transportin could be potentiated by AdOx treatment through modulating methylation status in RGG domains of FUS. These findings indicated the treatment with a methylation inhibitor at the appropriate levels could alleviate the cytoplasmic mislocalization but in excess this could cause the intranuclear aggregation of FUS C-terminal mutant.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2
Fig. 3

Similar content being viewed by others

References

  1. Vance C, Rogelj B, Hortobagyi T, De Vos KJ, Nishimura AL, Sreedharan J, Hu X, Smith B, Ruddy D, Wright P, Ganesalingam J, Williams KL, Tripathi V, Al-Saraj S, Al-Chalabi A, Leigh PN, Blair IP, Nicholson G, de Belleroche J, Gallo JM, Miller CC, Shaw CE (2009) Mutations in FUS, an RNA processing protein, cause familial amyotrophic lateral sclerosis type 6. Science 323:1208–1211

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Kwiatkowski TJ Jr, Bosco DA, Leclerc AL, Tamrazian E, Vanderburg CR, Russ C, Davis A, Gilchrist J, Kasarskis EJ, Munsat T, Valdmanis P, Rouleau GA, Hosler BA, Cortelli P, de Jong PJ, Yoshinaga Y, Haines JL, Pericak-Vance MA, Yan J, Ticozzi N, Siddique T, McKenna-Yasek D, Sapp PC, Horvitz HR, Landers JE, Brown RH Jr (2009) Mutations in the FUS/TLS gene on chromosome 16 cause familial amyotrophic lateral sclerosis. Science 323:1205–1208

    Article  CAS  PubMed  Google Scholar 

  3. Lagier-Tourenne C, Cleveland DW (2009) Rethinking ALS: the FUS about TDP-43. Cell 136:1001–1004

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Lagier-Tourenne C, Polymenidou M, Cleveland DW (2010) TDP-43 and FUS/TLS: emerging roles in RNA processing and neurodegeneration. Hum Mol Genet 19:R46–R64

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Dormann D, Rodde R, Edbauer D, Bentmann E, Fischer I, Hruscha A, Than ME, Mackenzie IR, Capell A, Schmid B, Neumann M, Haass C (2010) ALS-associated fused in sarcoma (FUS) mutations disrupt Transportin-mediated nuclear import. EMBO J 29:2841–2857

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Dormann D, Madl T, Valori CF, Bentmann E, Tahirovic S, Abou-Ajram C, Kremmer E, Ansorge O, Mackenzie IR, Neumann M, Haass C (2012) Arginine methylation next to the PY-NLS modulates Transportin binding and nuclear import of FUS. EMBO J 31:4258–4275

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Kato M, Han TW, Xie S, Shi K, Du X, Wu LC, Mirzaei H, Goldsmith EJ, Longgood J, Pei J, Grishin NV, Frantz DE, Schneider JW, Chen S, Li L, Sawaya MR, Eisenberg D, Tycko R, McKnight SL (2012) Cell-free formation of RNA granules: low complexity sequence domains form dynamic fibers within hydrogels. Cell 149:753–767

    Article  CAS  PubMed  Google Scholar 

  8. Kwon I, Kato M, Xiang S, Wu L, Theodoropoulos P, Mirzaei H, Han T, Xie S, Corden JL, McKnight SL (2013) Phosphorylation-regulated binding of RNA polymerase II to fibrous polymers of low-complexity domains. Cell 155:1049–1060

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Mackenzie IR, Rademakers R, Neumann M (2010) TDP-43 and FUS in amyotrophic lateral sclerosis and frontotemporal dementia. Lancet Neurol 9:995–1007

    Article  CAS  PubMed  Google Scholar 

  10. Du K, Arai S, Kawamura T, Matsushita A, Kurokawa R (2011) TLS and PRMT1 synergistically coactivate transcription at the survivin promoter through TLS arginine methylation. Biochem Biophys Res Commun 404:991–996

    Article  CAS  PubMed  Google Scholar 

  11. Tradewell ML, Yu Z, Tibshirani M, Boulanger MC, Durham HD, Richard S (2012) Arginine methylation by PRMT1 regulates nuclear-cytoplasmic localization and toxicity of FUS/TLS harbouring ALS-linked mutations. Hum Mol Genet 21:136–149

    Article  PubMed  Google Scholar 

  12. Yamaguchi A, Kitajo K (2012) The effect of PRMT1-mediated arginine methylation on the subcellular localization, stress granules, and detergent-insoluble aggregates of FUS/TLS. PLoS One 7:e49267

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Scaramuzzino C, Monaghan J, Milioto C, Lanson NA Jr, Maltare A, Aggarwal T, Casci I, Fackelmayer FO, Pennuto M, Pandey UB (2013) Protein arginine methyltransferase 1 and 8 interact with FUS to modify its sub-cellular distribution and toxicity in vitro and in vivo. PLoS One 8:e61576

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Lee HW, Kim S, Paik WK (1977) S-adenosylmethionine: protein-arginine methyltransferase. Purification and mechanism of the enzyme. Biochemistry 16:78–85

    Article  CAS  PubMed  Google Scholar 

  15. Nicholson TB, Chen T, Richard S (2009) The physiological and pathophysiological role of PRMT1-mediated protein arginine methylation. Pharmacol Res 60:466–474

    Article  CAS  PubMed  Google Scholar 

  16. Bedford MT, Clarke SG (2009) Protein arginine methylation in mammals: who, what, and why. Mol Cell 33:1–13

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Cheng D, Yadav N, King RW, Swanson MS, Weinstein EJ, Bedford MT (2004) Small molecule regulators of protein arginine methyltransferases. J Biol Chem 279:23892–23899

    Article  CAS  PubMed  Google Scholar 

  18. Wang J, Chen L, Sinha SH, Liang Z, Chai H, Muniyan S, Chou YW, Yang C, Yan L, Feng Y, Li KK, Lin MF, Jiang H, Zheng YG, Luo C (2012) Pharmacophore-based virtual screening and biological evaluation of small molecule inhibitors for protein arginine methylation. J Med Chem 55:7978–7987

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Williams-Ashman HG, Seidenfeld J, Galletti P (1982) Trends in the biochemical pharmacology of 5′-deoxy-5′-methylthioadenosine. Biochem Pharmacol 31:277–288

    Article  CAS  PubMed  Google Scholar 

  20. Osborne TC, Obianyo O, Zhang X, Cheng X, Thompson PR (2007) Protein arginine methyltransferase 1: positively charged residues in substrate peptides distal to the site of methylation are important for substrate binding and catalysis. Biochemistry 46:13370–13381

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Bonham K, Hemmers S, Lim YH, Hill DM, Finn MG, Mowen KA (2010) Effects of a novel arginine methyltransferase inhibitor on T-helper cell cytokine production. FEBS J 277:2096–2108

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Endo M, Ohashi K, Mizuno K (2007) LIM kinase and slingshot are critical for neurite extension. J Biol Chem 282:13692–13702

    Article  CAS  PubMed  Google Scholar 

  23. Koga S, Kojima S, Kishimoto T, Kuwabara S, Yamaguchi A (2012) Over-expression of map kinase phosphatase-1 (MKP-1) suppresses neuronal death through regulating JNK signaling in hypoxia/re-oxygenation. Brain Res 1436:137–146

    Article  CAS  PubMed  Google Scholar 

  24. Chen Y, Zhang L, Jones KA (2011) SKIP counteracts p53-mediated apoptosis via selective regulation of p21Cip1 mRNA splicing. Genes Dev 25:701–716

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Conte A, Lattante S, Zollino M, Marangi G, Luigetti M, Del Grande A, Servidei S, Trombetta F, Sabatelli M (2012) P525L FUS mutation is consistently associated with a severe form of juvenile amyotrophic lateral sclerosis. Neuromuscul Disord 22:73–75

    Article  PubMed  Google Scholar 

  26. Bosco DA, Lemay N, Ko HK, Zhou H, Burke C, Kwiatkowski TJ Jr, Sapp P, McKenna-Yasek D, Brown RH Jr, Hayward LJ (2010) Mutant FUS proteins that cause amyotrophic lateral sclerosis incorporate into stress granules. Hum Mol Genet 19:4160–4175

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Takanashi K, Yamaguchi A (2014) Aggregation of ALS-linked FUS mutant sequesters RNA binding proteins and impairs RNA granules formation. Biochem Biophys Res Commun 452:600–607

    Article  CAS  PubMed  Google Scholar 

  28. Boisvert FM, Cote J, Boulanger MC, Cleroux P, Bachand F, Autexier C, Richard S (2002) Symmetrical dimethylarginine methylation is required for the localization of SMN in Cajal bodies and pre-mRNA splicing. J Cell Biol 159:957–969

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Gal J, Zhang J, Kwinter DM, Zhai J, Jia H, Jia J, Zhu H (2011) Nuclear localization sequence of FUS and induction of stress granules by ALS mutants. Neurobiol Aging 32(12):2323.e27-40

    Article  PubMed  Google Scholar 

  30. Buratti E, Baralle FE (2012) TDP-43: gumming up neurons through protein-protein and protein-RNA interactions. Trends Biochem Sci 37:237–247

    Article  CAS  PubMed  Google Scholar 

  31. Zhou Y, Liu S, Liu G, Ozturk A, Hicks GG (2013) ALS-associated FUS mutations result in compromised FUS alternative splicing and autoregulation. PLoS Genet 9:e1003895

    Article  PubMed  PubMed Central  Google Scholar 

  32. Schwartz JC, Podell ER, Han SS, Berry JD, Eggan KC, Cech TR (2014) FUS is sequestered in nuclear aggregates in ALS patient fibroblasts. Mol Biol Cell 25:2571–2578

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgments

This work was supported by research grants from the Japan Society for the Promotion of Science (#25461267) and Japan ALS Association (JALSA).

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Atsushi Yamaguchi.

Ethics declarations

Conflict of interest

The authors declare that they have no competing interests.

Electronic supplementary material

Below is the link to the electronic supplementary material.

11064_2015_1758_MOESM1_ESM.mp4

HEK293 cells, transfected with GFP-FUS P525L plasmid, were treated with 60 μM AdOx for 24 h initiating at 12 h after transfection. Then cells were fixed, stained with DAPI, and observed under confocal laser scanning microscopy (Olympus FV10i). 3D animation of the nucleus was reconstructed using Olympus FV10i software (MP4 675 kb)

Supplementary Fig. 1

(A) HEK293 or SH-SY5Y cells, transfected with plasmid encoding HA-tagged FUS wt, FUS R521G, FUS P525L, or FUS dC, were fixed at 24 after transfection. Cells were immunostained with anti-HA antibody, and then incubated with DAPI. Images were obtained using fluorescence microscopy. Bars, 5 μm. (B) HEK293 cells, transfected with plasmids encoding GFP-FUS wt or each mutant, were lysed for Western blot analyses with anti-FUS and -Actin antibody at 24 h after transfection (upper panels). HEK293 cells, transfected with plasmid encoding GFP-FUS P525L, were lysed for Western blot analyses with anti-FUS and –Actin antibody at the indicated time points (lower panels). The bar graph indicates the relative fold of the band intensity for the ratio of ectopic GFP-FUS/endogenous FUS protein (n = 3, mean ± SE). (C) HEK293 cells, transiently transfected with GFP-FUS wt or HA-FUS wt plasmid, were treated with 60 μM AdOx for 24 h and then fixed with DAPI. Bars, 2 μm. Arrows indicate the intranuclear aggregates (PDF 506 kb)

Supplementary Fig. 2

(A) (B) HEK293 cells, transiently transfected with GFP-control (A) or HA-tagged FUS P525L (B), were treated with (+) or without (−) 20 μM AdOx for 24 h initiating at 12 h after transfection. Cells were fixed and then stained with DAPI. Images were obtained using fluorescence microscopy. Bars, 5 μm. (C) HEK293 and SH-SY5Y cells, transiently transfected with GFP-FUS P525L plasmid, were treated with AMI-1(0, 100, 400 μM) for 24 h initiating at 12 h after transfection. Cells were fixed and then stained with DAPI. Bars, 5 μm. (D) HEK293 cells were treated with Mock, 5 or 20 μM AdOx, or 800 μM MTA for 24 h (upper panel). HEK293 cells were also treated with Mock, 20 μM AdOx or 400 μM AMI-1for 24 h (lower panel). Then FUS proteins, obtained by the immunoprecipitation with anti-FUS antibody in lysates of HEK293 cells, were then resolved by SDS-PAGE. The asymmetric arginine-methylation levels were measured by the immunoblot with anti-dimethyl-Arginine, asymmetric (ASYM24) Antibody (Millipore, #07-414). IgG HC, IgG heavy chain. “Relative ratio” means the relative levels of methylation compared with those of Mock sample (value = 1) (PDF 276 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Fujii, S., Takanashi, K., Kitajo, K. et al. Treatment with a Global Methyltransferase Inhibitor Induces the Intranuclear Aggregation of ALS-Linked FUS Mutant In Vitro. Neurochem Res 41, 826–835 (2016). https://doi.org/10.1007/s11064-015-1758-z

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s11064-015-1758-z

Keywords

Navigation