Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Article
  • Published:

Interaction of FUS and HDAC1 regulates DNA damage response and repair in neurons

Abstract

Defects in DNA repair have been extensively linked to neurodegenerative diseases, but the exact mechanisms remain poorly understood. We found that FUS, an RNA/DNA-binding protein that has been linked to amyotrophic lateral sclerosis (ALS) and frontotemporal lobar degeneration, is important for the DNA damage response (DDR). The function of FUS in DDR involved a direct interaction with histone deacetylase 1 (HDAC1), and the recruitment of FUS to double-stranded break sites was important for proper DDR signaling. Notably, FUS proteins carrying familial ALS mutations were defective in DDR and DNA repair and showed a diminished interaction with HDAC1. Moreover, we observed increased DNA damage in human ALS patients harboring FUS mutations. Our findings suggest that an impaired DDR and DNA repair may contribute to the pathogenesis of neurodegenerative diseases linked to FUS mutations.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Figure 1: FUS is important for DDR and repair in proliferative cells and in postmitotic neurons.
Figure 2: FUS is rapidly recruited to DSBs and is one of the earliest proteins to respond to DNA damage.
Figure 3: FUS directly interacts with HDAC1 and both proteins are necessary for successful DNA repair.
Figure 4: The G-rich and C-terminal domains of FUS directly interact with HDAC1, and this interaction is important for successful DSB repair.
Figure 5: Cells expressing human fALS FUS mutations exhibit impaired DNA repair efficiency and a diminished FUS-HDAC1 interaction.
Figure 6: fALS patients harboring FUS mutations exhibit increased DNA damage.

Similar content being viewed by others

References

  1. Rass, U., Ahel, I. & West, S.C. Defective DNA repair and neurodegenerative disease. Cell 130, 991–1004 (2007).

    Article  CAS  PubMed  Google Scholar 

  2. McKinnon, P.J. DNA repair deficiency and neurological disease. Nat. Rev. Neurosci. 10, 100–112 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Bender, A. et al. High levels of mitochondrial DNA deletions in substantia nigra neurons in aging and Parkinson disease. Nat. Genet. 38, 515–517 (2006).

    Article  CAS  PubMed  Google Scholar 

  4. Lu, T. et al. Gene regulation and DNA damage in the ageing human brain. Nature 429, 883–891 (2004).

    Article  CAS  PubMed  Google Scholar 

  5. Anderson, A.J., Su, J.H. & Cotman, C.W. DNA damage and apoptosis in Alzheimer's disease: colocalization with c-Jun immunoreactivity, relationship to brain area, and effect of postmortem delay. J. Neurosci. 16, 1710–1719 (1996).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. de Waard, M.C. et al. Age-related motor neuron degeneration in DNA repair-deficient Ercc1 mice. Acta Neuropathol. 120, 461–475 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  7. Tan, A.Y. & Manley, J.L. The TET family of proteins: functions and roles in disease. J. Mol. Cell Biol. 1, 82–92 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Fujii, R. et al. The RNA-binding protein TLS is translocated to dendritic spines by mGluR5 activation and regulates spine morphology. Curr. Biol. 15, 587–593 (2005).

    Article  CAS  PubMed  Google Scholar 

  9. Fujii, R. & Takumi, T. TLS facilitates transport of mRNA encoding an actin-stabilizing protein to dendritic spines. J. Cell Sci. 118, 5755–5765 (2005).

    Article  CAS  PubMed  Google Scholar 

  10. Kuroda, M. et al. Male sterility and enhanced radiation sensitivity in TLS(−/−) mice. EMBO J. 19, 453–462 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Hicks, G.G. et al. Fus deficiency in mice results in defective B-lymphocyte development and activation, high levels of chromosomal instability and perinatal death. Nat. Genet. 24, 175–179 (2000).

    Article  CAS  PubMed  Google Scholar 

  12. Bertrand, P., Akhmedov, A.T., Delacote, F., Durrbach, A. & Lopez, B.S. Human POMp75 is identified as the pro-oncoprotein TLS/FUS: both POMp75 and POMp100 DNA homologous pairing activities are associated to cell proliferation. Oncogene 18, 4515–4521 (1999).

    Article  CAS  PubMed  Google Scholar 

  13. Baechtold, H. et al. Human 75-kDa DNA-pairing protein is identical to the pro-oncoprotein TLS/FUS and is able to promote D-loop formation. J. Biol. Chem. 274, 34337–34342 (1999).

    Article  CAS  PubMed  Google Scholar 

  14. Gardiner, M.T.R., Vandermoere, F., Morrice, N.A. & Rouse, J. Identification and characterization of FUS/TLS as a new target of ATM. Biochem. J. 415, 297–307 (2008).

    Article  CAS  PubMed  Google Scholar 

  15. Wang, X. et al. Induced ncRNAs allosterically modify RNA-binding proteins in cis to inhibit transcription. Nature 454, 126–130 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Vance, C. et al. Mutations in FUS, an RNA processing protein, cause familial amyotrophic lateral sclerosis type 6. Science 323, 1208–1211 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Kwiatkowski, T.J. Jr. et al. Mutations in the FUS/TLS gene on chromosome 16 cause familial amyotrophic lateral sclerosis. Science 323, 1205–1208 (2009).

    CAS  PubMed  Google Scholar 

  18. Pierce, A.J., Johnson, R.D., Thompson, L.H. & Jasin, M. XRCC3 promotes homology-directed repair of DNA damage in mammalian cells. Genes Dev. 13, 2633–2638 (1999).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Patel, K.J. et al. Involvement of Brca2 in DNA repair. Mol. Cell 1, 347–357 (1998).

    Article  CAS  PubMed  Google Scholar 

  20. Seluanov, A., Mittelman, D., Pereira-Smith, O.M., Wilson, J.H. & Gorbunova, V. DNA end joining becomes less efficient and more error-prone during cellular senescence. Proc. Natl. Acad. Sci. USA 101, 7624–7629 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Ferguson, D.O. et al. The nonhomologous end-joining pathway of DNA repair is required for genomic stability and the suppression of translocations. Proc. Natl. Acad. Sci. USA 97, 6630–6633 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Sharma, S. Age-related nonhomologous end joining activity in rat neurons. Brain Res. Bull. 73, 48–54 (2007).

    Article  PubMed  Google Scholar 

  23. Hande, K.R. Etoposide: four decades of development of a topoisomerase II inhibitor. Eur. J. Cancer 34, 1514–1521 (1998).

    Article  CAS  PubMed  Google Scholar 

  24. Fillingham, J., Keogh, M.C. & Krogan, N.J. GammaH2AX and its role in DNA double-strand break repair. Biochem. Cell Biol. 84, 568–577 (2006).

    Article  CAS  PubMed  Google Scholar 

  25. Anderson, L., Henderson, C. & Adachi, Y. Phosphorylation and rapid relocalization of 53BP1 to nuclear foci upon DNA damage. Mol. Cell Biol. 21, 1719–1729 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Keramaris, E., Hirao, A., Slack, R.S., Mak, T.W. & Park, D.S. Ataxia telangiectasia–mutated protein can regulate p53 and neuronal death independent of Chk2 in response to DNA damage. J. Biol. Chem. 278, 37782–37789 (2003).

    Article  CAS  PubMed  Google Scholar 

  27. Kim, D. et al. Deregulation of HDAC1 by p25/Cdk5 in neurotoxicity. Neuron 60, 803–817 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Collins, A.R. The comet assay for DNA damage and repair: principles, applications and limitations. Mol. Biotechnol. 26, 249–261 (2004).

    Article  CAS  PubMed  Google Scholar 

  29. Pilch, D.R. et al. Characteristics of gamma-H2AX foci at DNA double-strand breaks sites. Biochem. Cell Biol. 81, 123–129 (2003).

    Article  CAS  PubMed  Google Scholar 

  30. Soutoglou, E. & Misteli, T. Activation of the cellular DNA damage response in the absence of DNA lesions. Science 320, 1507–1510 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Weinstock, D.M., Nakanishi, K., Helgadottir, H.R. & Jasin, M. Assaying double-strand break repair pathway choice in mammalian cells using a targeted endonuclease or the RAG recombinase. Methods Enzymol. 409, 524–540 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Dobbin, M.M. et al. SIRT1 collaborates with ATM and HDAC1 to maintain genomic stability in neurons. Nat. Neurosci. 16, 1008–1015 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Stark, J.M. et al. ATP hydrolysis by mammalian RAD51 has a key role during homology-directed DNA repair. J. Biol. Chem. 277, 20185–20194 (2002).

    Article  CAS  PubMed  Google Scholar 

  34. Huang, E.J. et al. Extensive FUS-immunoreactive pathology in juvenile amyotrophic lateral sclerosis with basophilic inclusions. Brain Pathol. 20, 1069–1076 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Xu, Y. et al. Targeted disruption of ATM leads to growth retardation, chromosomal fragmentation during meiosis, immune defects, and thymic lymphoma. Genes Dev. 10, 2411–2422 (1996).

    Article  CAS  PubMed  Google Scholar 

  36. Celeste, A. et al. Genomic instability in mice lacking histone H2AX. Science 296, 922–927 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Miller, K.M. et al. Human HDAC1 and HDAC2 function in the DNA-damage response to promote DNA nonhomologous end-joining. Nat. Struct. Mol. Biol. 17, 1144–1151 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Pegoraro, G. et al. Ageing-related chromatin defects through loss of the NURD complex. Nat. Cell Biol. 11, 1261–1267 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Lukas, J., Lukas, C. & Bartek, J. More than just a focus: The chromatin response to DNA damage and its role in genome integrity maintenance. Nat. Cell Biol. 13, 1161–1169 (2011).

    Article  CAS  PubMed  Google Scholar 

  40. Dinant, C., Houtsmuller, A.B. & Vermeulen, W. Chromatin structure and DNA damage repair. Epigenetics Chromatin. 1, 9 (2008).

    Article  PubMed  PubMed Central  Google Scholar 

  41. Guan, J.S. et al. HDAC2 negatively regulates memory formation and synaptic plasticity. Nature 459, 55–60 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Gräff, J. et al. An epigenetic blockade of cognitive functions in the neurodegenerating brain. Nature 483, 222–226 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  43. Crozat, A., Aman, P., Mandahl, N. & Ron, D. Fusion of CHOP to a novel RNA-binding protein in human myxoid liposarcoma. Nature 363, 640–644 (1993).

    Article  CAS  PubMed  Google Scholar 

  44. Ferrante, R.J. et al. Evidence of increased oxidative damage in both sporadic and familial amyotrophic lateral sclerosis. J. Neurochem. 69, 2064–2074 (1997).

    Article  CAS  PubMed  Google Scholar 

  45. Millecamps, S. et al. Phenotype difference between ALS patients with expanded repeats in C9ORF72 and patients with mutations in other ALS-related genes. J. Med. Genet. 49, 258–263 (2012).

    Article  CAS  PubMed  Google Scholar 

  46. Huang, E.J. et al. Extensive FUS-immunoreactive pathology in juvenile amyotrophic lateral sclerosis with basophilic inclusions. Brain Pathol. 20, 1069–1076 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Lois, C., Hong, E.J., Pease, S., Brown, E.J. & Baltimore, D. Germline transmission and tissue-specific expression of transgenes delivered by lentiviral vectors. Science 295, 868–872 (2002).

    Article  CAS  PubMed  Google Scholar 

  48. Wang, W. et al. Genetically encoding unnatural amino acids for cellular and neuronal studies. Nat. Neurosci. 10, 1063–1072 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Gao, J. et al. A novel pathway regulates memory and plasticity via SIRT1 and miR-134. Nature 466, 1105–1109 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  50. Kruhlak, M.J., Celeste, A. & Nussenzweig, A. Monitoring DNA breaks in optically highlighted chromatin in living cells by laser scanning confocal microscopy. Methods Mol. Biol. 523, 125–140 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Carpenter, A.E. et al. CellProfiler: image analysis software for identifying and quantifying cell phenotypes. Genome Biol 7, R100 (2006).

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank T. Misteli (National Cancer Institute) for kindly providing the NIH2/4 cell line and Lac-R constructs, M. Dobbin (Massachusetts Institute of Technology) for technical assistance on microirradiation experiments, A. Mungenast, R. Madabhushi, A. Bero, J. Penney and A. Nott for critical reading of the manuscript, and other members of the Tsai and Huang laboratories for helpful discussions. We also thank R.H. Brown Jr and L.J. Hayward for insightful discussions. W.-Y.W. was supported by a Postdoctoral Fellowship from the Simons Foundation, J.C.J. was supported by the Howard Hughes Medical Institute Exceptional Research Opportunities Program. This work is also supported by grants from National Institute of Neurological Disorders and Stroke (NS078839 to L.-H.T.), the Department of Veterans Affairs (BX001108 to E.J.H.) and the Muscular Dystrophy Association (MDA217592 to E.J.H.). L.-H.T. is a member of the Neurodegeneration Consortium. E.J.H. is a member of the Consortium for Frontotemporal Dementia Research. L.-H.T. receives funding from the Howard Hughes Medical Institute.

Author information

Authors and Affiliations

Authors

Contributions

W.-Y.W. performed the DNA repair assay, in vitro protein interaction, domain mapping assay, created mutant cell lines, and completed the immunoprecipitation, ChIP and qPCR assays. L.P. performed the micro-irradiation and comet assays and evaluated DNA damage response in cultured neurons. S.C.S. purified full-length and fragment GST-tagged proteins. E.J.Q. and M.S. helped with neuronal culture. J.C.J. helped with the in vitro GST pulldown assay. E.J.H. and I.R.A.M. provided brain sections from control individuals and human fALS patients. L.-H.T. and E.J.H. supervised the project, and W.-Y.W. and L.-H.T. wrote the manuscript.

Corresponding author

Correspondence to Li-Huei Tsai.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 Schematic figures of HR and NHEJ reporter systems used in this study.

(a). The I-SceI mediated DSB induction and repair by HR. A I-SceI endonuclease recognition site was inserted into the GFP coding sequence to create a nonfunctional GFP transgene, and a wild type GFP fragment was inserted downstream of the GFP tansgene. Transfection of I-SceI expressing construct will induce a DSB at the I-SecI site, and repair of this DSB by HR will successfully restore a functional GFP, which could be detected by fluorescence microscope or by FACS. Arrows in up panel (red) represent the primers used for ChIP-qPCR analysis of the repair proteins accumulted at DSBs. (b). Reporter construct for NHEJ. The GFP coding sequence was interrupted by inserting an adenoviral exon flanked by artificial introns. The GFP is inactive due to the expression of the adeno exon in GFP gene; however, once digested with HindIII or I-SceI, the exon will be cutted out and a DSB will be created. Successful repair of this DSB using NHEJ will restore the expression of GFP. (Adapted from Oberdoerffer et al, cell, 2008; Seluanov et. al. PNAS. 2004).

Supplementary Figure 2 Verification of the shRNAs and siRNA used in this study.

(a-c). Different siRNAs targeting human BRCA2, LIG4, FUS and shRNAs targeting mouse Fus (shRNA2 and 3) or human FUS (shRNA 2, 4-6) were transfected into 293T (a,c) cells and N2A cells (b) for 72 hours and cell lysate was collected for western blotting.

Supplementary Figure 3 Immunofluorescence staining of 53BP1 and γH2AX in vehicle treated neurons following Fus knockdown.

(a). Primary cortical neurons were transfected with plasmids expressing scrambled shRNA or Fus shRNAs together with mCherry, treated with vechile for 1 hour fixed and labeled for yH2AX. No yH2AX immunoreactivity was observed in both transfected (white arrows) and non-transfected neurons. scale bar: 8μm (b). Primary cortical neurons were transduced with lentivirus carrying shRNAs targeting Fus, or scrambled control shRNA. Neurons were treated with vehicle for 1 h and labeled with anti-53BP1 antibody. 53BP1 is uniformly distributed in the nuclei. Scale bar: 4μm

Supplementary Figure 4 Immunofluorescence staining of phospho-ChK2 (p-ChK2) in cultured primary neurons treated with ETO.

(a) Primary cortical neurons were transfected with plasmids expressing scrambled shRNA or Fus shRNAs together with mCherry. 5 AM etoposide was added to induce DNA DSBs. Following 1 h etoposide treatment, neurons were fixed and labeled for p-ChK2 immunoreactivity. Fus knockdown neurons show reduced p-ChK2 immunoreactivity compared to scrambled shRNA-expressing neurons (white arrows). (b) p-ChK2 signal intensity in transfected neurons was measured by ImageJ (NIH). Scale bar: 8um. (** p<0.01, one-way ANOVA). (c). Verification of p-ChK2 antibody used for the immunofluorescnence staining.The p-ChK2 antibody (Cell Signaling #2661) detects a major band of approximately 62 kD. When the blot was stripped and re-blotted with ChK2 antibody (Cell Signaling #2662), the same 62 kD species was recognized. The p-ChK2 signal was minimal in vehicle-treated neurons and was enhanced with etoposide treatment, whereas phosphatase treatment completely abolished the signal. Note that the ChK2 antibody (Cell Signaling #2662) can recognize both human and mouse ChK2.

Supplementary Figure 5 In vivo interaction between HDAC and FUS.

Nuclear extracts prepared from WT mouse hippocampus were immunoprecipitated with antibodies against HDAC1 and HDAC2 and blotted with anti-FUS antibody.

Supplementary Figure 6 Micro-irradiation assay for HDAC1 and γH2AX.

(a). lmmunolabeling for HDAC1 and yH2AX was conducted at the indicated times following the induction of DSBs via laser micro-irradiation of U20S cells. Scale bar: 8um. (b). Fluorescence intensity in the laser-irradiated area at each time points was first normalized to the signal of whole nucleus and then to the peak fluorescence intentsity accross time. (n=5-10)

Supplementary Figure 7 Input controls for the experiment depicted in Figure 4b.

Flag tagged FUS fragments (FG) 4, 5, 7, or 4+7 were transfected into 293T cells together with FUS-mCherry for 48 hr and processed for immunoprecipitation as shown in Figure 4b. This figure shows the input controls for these experiments.

Supplementary Figure 8 Cellular distribution of wild type and mutant FUS.

Primary cultured neurons were transfected with FUS-WT, FUS-R244C, FUS-R514S, FUS-H517Q, or FUS-R521C, and the percentage of the cells showing a cytoplasmic accumulation of FUS was analyzed for each condition (n ≥ 50, ns: no significant difference. ***p<0.001, unpaired t-test). Scale bar: 6μm

Supplementary Figure 9 Laser micro-irradiation assay in U2OS cells expressing wild type FUS or fALS FUS mutants.

Endogenous FUS was knocked down and replaced by mCherry tagged wild type or mutant FUS. Cells were fixed 10 minutes after laser irradiation and processed for immunochemistry with anti-mCherry antibody.Scale bar: 4μm.

Supplementary Figure 10 Full-length pictures of the blots presented in the main figures.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–10 and Supplementary Table 1 (PDF 26595 kb)

Rights and permissions

Reprints and permissions

About this article

Cite this article

Wang, WY., Pan, L., Su, S. et al. Interaction of FUS and HDAC1 regulates DNA damage response and repair in neurons. Nat Neurosci 16, 1383–1391 (2013). https://doi.org/10.1038/nn.3514

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nn.3514

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing