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.

GGGGCC repeat expansion in C9orf72 compromises nucleocytoplasmic transport

Abstract

The GGGGCC (G4C2) repeat expansion in a noncoding region of C9orf72 is the most common cause of sporadic and familial forms of amyotrophic lateral sclerosis and frontotemporal dementia1,2. The basis for pathogenesis is unknown. To elucidate the consequences of G4C2 repeat expansion in a tractable genetic system, we generated transgenic fly lines expressing 8, 28 or 58 G4C2-repeat-containing transcripts that do not have a translation start site (AUG) but contain an open-reading frame for green fluorescent protein to detect repeat-associated non-AUG (RAN) translation. We show that these transgenic animals display dosage-dependent, repeat-length-dependent degeneration in neuronal tissues and RAN translation of dipeptide repeat (DPR) proteins, as observed in patients with C9orf72-related disease. This model was used in a large-scale, unbiased genetic screen, ultimately leading to the identification of 18 genetic modifiers that encode components of the nuclear pore complex (NPC), as well as the machinery that coordinates the export of nuclear RNA and the import of nuclear proteins. Consistent with these results, we found morphological abnormalities in the architecture of the nuclear envelope in cells expressing expanded G4C2 repeats in vitro and in vivo. Moreover, we identified a substantial defect in RNA export resulting in retention of RNA in the nuclei of Drosophila cells expressing expanded G4C2 repeats and also in mammalian cells, including aged induced pluripotent stem-cell-derived neurons from patients with C9orf72-related disease. These studies show that a primary consequence of G4C2 repeat expansion is the compromise of nucleocytoplasmic transport through the nuclear pore, revealing a novel mechanism of neurodegeneration.

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

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

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

Figure 1: G4C2 repeats induces length- and dosage-dependent degeneration in Drosophila.
Figure 2: Genetic screen identifies multiple modifiers of (G4C2)58 toxicity in the nucleocytoplasmic transport pathway.
Figure 3: Drosophila salivary gland cells expressing (G4C2)58 exhibit nuclear envelope abnormalities and accumulation of nuclear RNA.
Figure 4: Accumulation of nuclear RNA in human cells expressing G4C2 repeat expansion.

Similar content being viewed by others

References

  1. DeJesus-Hernandez, M. et al. Expanded GGGGCC hexanucleotide repeat in noncoding region of C9ORF72 causes chromosome 9p-linked FTD and ALS. Neuron 72, 245–256 (2011)

    Article  CAS  Google Scholar 

  2. Renton, A. E. et al. A hexanucleotide repeat expansion in C9ORF72 is the cause of chromosome 9p21-linked ALS-FTD. Neuron 72, 257–268 (2011)

    Article  CAS  Google Scholar 

  3. Mizielinska, S. et al. C9orf72 repeat expansions cause neurodegeneration in Drosophila through arginine-rich proteins. Science 345, 1192–1194 (2014)

    Article  ADS  CAS  Google Scholar 

  4. Xu, Z. et al. Expanded GGGGCC repeat RNA associated with amyotrophic lateral sclerosis and frontotemporal dementia causes neurodegeneration. Proc. Natl Acad. Sci. USA 110, 7778–7783 (2013)

    Article  ADS  CAS  Google Scholar 

  5. Kwon, I. et al. Poly-dipeptides encoded by the C9orf72 repeats bind nucleoli, impede RNA biogenesis, and kill cells. Science 345, 1139–1145 (2014)

    Article  ADS  CAS  Google Scholar 

  6. Wen, X. et al. Antisense proline-arginine RAN dipeptides linked to C9ORF72-ALS/FTD form toxic nuclear aggregates that initiate in vitro and in vivo neuronal death. Neuron 84, 1213–1225 (2014)

    Article  CAS  Google Scholar 

  7. Makise, M. et al. The Nup153-Nup50 protein interface and its role in nuclear import. J. Biol. Chem. 287, 38515–38522 (2012)

    Article  CAS  Google Scholar 

  8. Cesario, J. & McKim, K. S. RanGTP is required for meiotic spindle organization and the initiation of embryonic development in Drosophila . J. Cell Sci. 124, 3797–3810 (2011)

    Article  CAS  Google Scholar 

  9. Reed, R. Coupling transcription, splicing and mRNA export. Curr. Opin. Cell Biol. 15, 326–331 (2003)

    Article  CAS  Google Scholar 

  10. Viphakone, N. et al. TREX exposes the RNA-binding domain of Nxf1 to enable mRNA export. Nat. Commun. 3, 1006 (2012)

    Article  ADS  Google Scholar 

  11. Zhou, Z. et al. The protein Aly links pre-messenger-RNA splicing to nuclear export in metazoans. Nature 407, 401–405 (2000)

    Article  ADS  CAS  Google Scholar 

  12. Chang, C. T. et al. Chtop is a component of the dynamic TREX mRNA export complex. EMBO J. 32, 473–486 (2013)

    Article  CAS  Google Scholar 

  13. Wan, J. et al. Mutations in the RNA exosome component gene EXOSC3 cause pontocerebellar hypoplasia and spinal motor neuron degeneration. Nature Genet. 44, 704–708 (2012)

    Article  CAS  Google Scholar 

  14. Cheng, H. et al. Human mRNA export machinery recruited to the 5′ end of mRNA. Cell 127, 1389–1400 (2006)

    Article  CAS  Google Scholar 

  15. Boehmer, T., Enninga, J., Dales, S., Blobel, G. & Zhong, H. Depletion of a single nucleoporin, Nup107, prevents the assembly of a subset of nucleoporins into the nuclear pore complex. Proc. Natl Acad. Sci. USA 100, 981–985 (2003)

    Article  ADS  CAS  Google Scholar 

  16. Vasu, S. et al. Novel vertebrate nucleoporins Nup133 and Nup160 play a role in mRNA export. J. Cell Biol. 155, 339–354 (2001)

    Article  CAS  Google Scholar 

  17. Walther, T. C. et al. The conserved Nup107–160 complex is critical for nuclear pore complex assembly. Cell 113, 195–206 (2003)

    Article  CAS  Google Scholar 

  18. Murphy, R. & Wente, S. R. An RNA-export mediator with an essential nuclear export signal. Nature 383, 357–360 (1996)

    Article  ADS  CAS  Google Scholar 

  19. Nousiainen, H. O. et al. Mutations in mRNA export mediator GLE1 result in a fetal motoneuron disease. Nature Genet. 40, 155–157 (2008)

    Article  CAS  Google Scholar 

  20. Kaneb, H. M. et al. Deleterious mutations in the essential mRNA metabolism factor, hGle1, in amyotrophic lateral sclerosis. Hum. Mol. Genet. 24, 1363–1373 (2015)

    Article  CAS  Google Scholar 

  21. Han, S. S., Williams, L. A. & Eggan, K. C. Constructing and deconstructing stem cell models of neurological disease. Neuron 70, 626–644 (2011)

    Article  CAS  Google Scholar 

  22. van Blitterswijk, M., DeJesus-Hernandez, M. & Rademakers, R. How do C9ORF72 repeat expansions cause amyotrophic lateral sclerosis and frontotemporal dementia: can we learn from other noncoding repeat expansion disorders? Curr. Opin. Neurol. 25, 689–700 (2012)

    Article  CAS  Google Scholar 

  23. Almeida, S. et al. Modeling key pathological features of frontotemporal dementia with C9ORF72 repeat expansion in iPSC-derived human neurons. Acta Neuropathol. 126, 385–399 (2013)

    Article  CAS  Google Scholar 

  24. Zhang, Z. et al. Downregulation of microRNA-9 in iPSC-derived neurons of FTD/ALS patients with TDP-43 mutations. PLoS ONE 8, e76055 (2013)

    Article  ADS  CAS  Google Scholar 

  25. Sareen, D. et al. Targeting RNA foci in iPSC-derived motor neurons from ALS patients with a C9ORF72 repeat expansion. Sci. Transl. Med. 5, 208ra149 (2013)

    Article  Google Scholar 

  26. Cronshaw, J. M. & Matunis, M. J. The nuclear pore complex: disease associations and functional correlations. Trends Endocrinol. Metab. 15, 34–39 (2004)

    Article  CAS  Google Scholar 

  27. Savas, J. N., Toyama, B. H., Xu, T., Yates, J. R., III & Hetzer, M. W. Extremely long-lived nuclear pore proteins in the rat brain. Science 335, 942 (2012)

    Article  ADS  CAS  Google Scholar 

  28. Mor, A., White, M. A. & Fontoura, B. M. Nuclear trafficking in health and disease. Curr. Opin. Cell Biol. 28, 28–35 (2014)

    Article  CAS  Google Scholar 

  29. Bonnet, A. & Palancade, B. Regulation of mRNA trafficking by nuclear pore complexes. Genes 5, 767–791 (2014)

    Article  Google Scholar 

  30. Zhang, Y. J. et al. Aggregation-prone c9FTD/ALS poly(GA) RAN-translated proteins cause neurotoxicity by inducing ER stress. Acta Neuropathol. 128, 505–524 (2014)

    Article  CAS  Google Scholar 

  31. Kim, N. C. et al. VCP is essential for mitochondrial quality control by PINK1/Parkin and this function is impaired by VCP mutations. Neuron 78, 65–80 (2013)

    Article  CAS  Google Scholar 

  32. Smith, R. & Taylor, J. P. Dissection and imaging of active zones in the Drosophila neuromuscular junction. J. Vis. Exp. (2011)

  33. Gendron, T. F. et al. Antisense transcripts of the expanded C9ORF72 hexanucleotide repeat form nuclear RNA foci and undergo repeat-associated non-ATG translation in c9FTD/ALS. Acta Neuropathol. 126, 829–844 (2013)

    Article  CAS  Google Scholar 

  34. Ash, P. E. et al. Unconventional translation of C9ORF72 GGGGCC expansion generates insoluble polypeptides specific to c9FTD/ALS. Neuron 77, 639–646 (2013)

    Article  CAS  Google Scholar 

  35. Almeida, S. et al. Induced pluripotent stem cell models of progranulin-deficient frontotemporal dementia uncover specific reversible neuronal defects. Cell Rep 2, 789–798 (2012)

    Article  CAS  Google Scholar 

  36. Okita, K. et al. A more efficient method to generate integration-free human iPS cells. Nature Methods 8, 409–412 (2011)

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank the Bloomington Drosophila Stock Center, the VDRC Stock Center, K. McKim and E. Baehrecke for fly lines, V. Budnik for Lamin C antibody, as well as the Cell and Tissue Imaging Center at St Jude Children’s Research Hospital and the University of Massachusetts Medical School Confocal Core for assistance. This work was supported by grants from Target ALS, The Packard Center for ALS Research at the Johns Hopkins University, and the ALS Association to F.-B.G., and J.P.T., and ALS Therapy Alliance, NIH (N079725) to F.-B.G., NIH (NS079725 and AG019724) to B.L.M., and the American-Lebanese-Syrian Associated Charities to J.P.T.

Author information

Authors and Affiliations

Authors

Contributions

F.-B.G. and J.P.T. conceived and supervised the project. B.D.F., Y.L., H.J.K., F.-B.G. and J.P.T. wrote the manuscript. B.D.F. and Y.L. performed the genetic screen and validation. B.D.F., Y.L., N.C.K., N.B. and K.-H.L. characterized Drosophila phenotypes and performed the assays characterizing RNA export in human cells. S.A. established human fibroblast cell lines, R.L.-G. generated some iPSC lines; S.A. and R.L.-G. performed cortical neuron differentiation. M.V. and B.D.F. conducted FISH experiments. L.P., B.L.M. and P.C.W. provided key reagents.

Corresponding authors

Correspondence to Fen-Biao Gao or J. Paul Taylor.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Extended data figures and tables

Extended Data Figure 1 Expression of G4C2 repeats induces length-dependent phenotypes in Drosophila.

a, Expression of (G4C2)58 in Drosophila motor neurons using the OK371‐GAL4 driver leads to a significant reduction in active zones as immunostained by the anti‐Bruchpilot antibody NC82 and anti-HRP. Scale bar, 50 μm. b, Quantification of active zones (n = 6 individual larvae for control, 4 for 2× (G4C2)8, and 6 for 2× (G4C2)58). Values are mean ± s.e.m. ** P < 0.01, one-way ANOVA, Tukey’s post hoc test. c, Pan neuronal expression of (G4C2)58 repeats induces dosage-dependent decrease in larval size (left) and locomotor activity measured in 30 s (right) when (G4C2)58 is expressed in all neurons using the elav-GAL4 driver. d, Quantification of the distance travelled by third instar larvae reveals expressing two copies of (G4C2)58 results in a significant deficit in locomotor activity. Values are mean ± s.e.m. (n = 7 individual larvae for control, 4 for 1× (G4C2)58, and 5 for 2′ (G4C2)58). **P < 0.01, one-way ANOVA, Tukey’s post hoc test. e, Pan neuronal expression of (G4C2)58 repeats in Drosophila neurons using the elav-GAL4 driver leads to a significant reduction in the bouton number. Bouton number was quantified by examining the presynaptic (anti-HRP) and postsynaptic (anti-DLG1) markers (left). Scale bar, 50 μm. f, Quantification of bouton number (left) and muscle size (right) reveal that both are significantly reduced in Drosophila larvae expressing (G4C2)58 repeats. Values are mean ± s.e.m. (n = 6 individual larvae for control, 5 for 2× (G4C2)8, and 6 for 2× (G4C2)58), ** P < 0.01, one-way ANOVA, Tukey’s post hoc test. g, Expression of (G4C2)28 and (G4C2)58 but not (G4C2)8 in the muscle using the MHC-GAL4 driver leads to loss of wing control in adult flies (n = 30 individual Drosophila for control, 21 for (G4C2)8, 50 for (G4C2)28, and 24 for (G4C2)58). This phenotype was assessed by examining the permanent wing posture of live adult flies.

Extended Data Figure 2 RAN translation is observed in Drosophila expressing G4C2 repeats.

a, Western blot revealing translation of RAN poly-dipeptides in flies expressing (G4C2)58 in the eye. RAN poly-dipeptides were not found in flies expressing (G4C2)8 or control flies. There was minimal expression of GFP-positive product observed in flies expressing (G4C2)28. GFP-expressing flies (lane 1) were used as a positive control for the anti-GFP antibody. b, Western blot showing production of RAN product when (G4C2)28 and (G4C2)58 but not (G4C2)8 repeats are expressed in the muscle. RAN products were visualized with anti-GFP antibody (left) and anti-poly(GP) antibody (right). c, The RAN product poly-GP–GFP from flies expressing (G4C2)58 in the muscle form large visible inclusions as visualized under light sheet fluorescent microscopy (left) and by confocal microscopy (right). Scale bar, 50 μm. d, Expression of (G4C2)58 in the salivary gland cells results in the formation of large nuclear inclusions and smaller cytoplasmic inclusions. Scale bar, 50 μm. e, f, Expression of (G4C2)58 in the ventral ganglion by OK371 driver results in the formation of nuclear and cytoplasmic inclusions, whereas GFP shows diffused nuclear and cytoplasmic localization. Lamin staining shows nuclear membrane, and CD8–RFP shows plasma membrane. Scale bar, 10 μm. g, Expression of (G4C2)58 in pan neuronal cells by elav driver results in the nuclear and cytoplasmic inclusions. Scale bar, 10 μm.

Extended Data Figure 3 Ectopic expression of poly(GR) but not poly(GA) or poly(GP) peptides are toxic in Drosophila.

a, Transgenic Drosophila were generated that express ATG-driven poly(GA), poly(GR) and poly(GP) peptides with an N-terminal GFP tag (top). Expression of GFP–(GA)50 and GFP–(GP)47 were non-toxic when expressed in the eye with GMR-GAL4 whereas GFP–(GR)50 expression resulted in >95% lethality with surviving adults having severely degenerated eyes (bottom). b, Western blot showing the expression of (G4C2)58, GFP–(GA)50, GFP–(GR)50 and GFP–(GP)47 as visualized in muscle by anti-GFP antibody. c, Western blot showing the expression of poly(GP) in muscle of flies expressing (G4C2)58 and GFP–(GP)47 but not GFP–(GA)50, GFP–(GR)50 and control flies as visualized by anti-GP antibody. d, Dot blot analysis of RAN peptides in muscle revealing expression of poly(GA) only in GFP–(GA)50 flies, expression of poly(GR) in (G4C2)58 and GFP–(GR)50 flies. As expected, anti-sense DPR poly(PR) was not found in any of the lysates. The background protein signal was used as a loading control.

Extended Data Figure 4 Nuclear import and export is altered by (G4C2)58 expression.

a, A threonine to asparagine substitution at residue 24 in the Ran protein abolishes the affinity for GTP and reduces its affinity for GDP. Hence, the RanT24N is always in either a nucleotide-free state or in its inactive, GDP-bound state, and acts as dominant negative. RANT24N expression driven by GMR-GAL4 causes a mild eye phenotype when expressed in the absence of (G4C2)58 (upper row, right panel). The (G4C2)58 rough eye phenotype is strongly enhanced by dominant-negative RanT24N expression (middle row, left panel). The (G4C2)58 eye phenotype is strongly enhanced by knockdown of Nup153 by two independent RNAi lines (middle row, two right panels). The (G4C2)58 eye phenotype is also mildly enhanced by knockdown of transportin (Trn) (bottom row). b, Knockdown of Crm1 in flies expressing (G4C2)58 induces a mild enhancement of the (G4C2)58 eye phenotype (left versus middle). Crm1 knockdown in the absence of (G4C2)58 repeats does not produce a rough eye phenotype (right). c, Expression of two copies of (G4C2)58 in the Drosophila motor neurons leads to reduced viability (50%). Chemical inhibition of Crm1 with Leptomycin B (500 nM) enhances (G4C2)58 toxicity resulting in reduced viability (23%). Leptomycin B does not impede viability (100%) in Drosophila expressing GFP. n is displayed on the graph and represents the individual pupae from two separate experiments.

Extended Data Figure 5 Phenotypes of additional suppressors and enhancers of (G4C2)58.

a, Phenotypes demonstrating suppression of the (G4C2)58 rough eye phenotype by RNAi knockdown of identified genes. b, Phenotypes demonstrating enhancement of the (G4C2)58 rough eye phenotype by RNAi knockdown of identified genes. c, Knockdown of identified modifier genes shows little or no phenotype in the absence of G4C2 repeat expression.

Extended Data Figure 6 Impairment of nucleocytoplasmic shuttling in Drosophila and cultured human cell lines.

a, (G4C2)58 expression driven by Fkh-GAL4 causes an abnormal nuclear envelope as shown by Lamin C staining (bottom) in comparison to (G4C2)8 (top). Scale bar, 10 μm. b, Transfection of 293T cells with (G4C2)58 (bottom) but not (G4C2)8 (top) leads to an increase in nuclear RNA puncta as visualized with a total RNA-FISH probe. Non-transfected cells (absence of GFP signal) do not show an increase in nuclear RNA in either (G4C2)8 or (G4C2)58 transfected cells. Scale bar, 25 μm. c, Enlarged images showing slowed accumulation of newly synthesized RNA in the cytoplasm of HeLa cells expressing (G4C2)58. Scale bar, 25 μm.

Extended Data Figure 7 Characterization of newly generated integration-free iPSC lines.

a, iPSC lines from a control subject (line 11) and a G4C2 repeat expansion carrier (line 3 and line 8) express pluripotent markers SSEA-4, Nanog and Oct-4. Scale bar, 50 μm. b, qRT–PCR analysis of expression levels of pluripotent stem-cell markers SOX2 and Nanog in these iPSC lines showing no statistical differences between these lines and human embryonic stem-cell line H9. Relative mRNA levels are quantified from 3 independent experiments. c, After differentiation into cortical neurons about 90% of cells in these cultures are MAP2-positive neurons. Scale bar, 50 μm. d, Quantification of average percentage of MAP2-positive neurons and there is no difference between control and C9orf72 cultures. Average percentages were quantified from 3 independent experiments. e, Quantification of average percentage of VGLUT-positive excitatory neurons among all neurons; there is no difference between control and C9orf72 cultures. n = 3 independent experiments.

Extended Data Figure 8 Karyotyping analysis and pluripotency of newly generated iPSC lines.

a, G-band staining showing a normal karyotype for all the lines analysed. b, After in vitro spontaneous differentiation of control and C9 carrier iPSC lines, cells were stained for α-fetoprotein (endoderm), desmin (mesoderm), βIII-tubulin (ectoderm) and Hoechst (nuclei). All lines showed differentiation towards derivates of three germ layers. Scale bars, 20 μm.

Extended Data Figure 9 Accumulation of nuclear RNA is not seen in fibroblasts derived from patients with G4C2 repeat expansion.

a, b, Total cellular RNA was measured by FISH in fibroblasts derived from 4 control (a) subjects or 5 patients (b) with G4C2 repeat expansion. Scale bar, 25 μm. c, Quantification shows no statistical difference in the observed nuclear to cytoplasmic RNA ratio in patient versus control fibroblasts. n = 16 individual cells analysed for each line.

Extended Data Figure 10 qRT–PCR analysis.

af, qRT–PCR analysis demonstrating knockdown of selected modifiers in Drosophila eyes. mRNA levels of selected modifier (a, d), GAL4 (b, e) and (G4C2)58 (c, f) assayed by qRT–PCR in progeny resulting from wild type (w1118), classical mutant allele or UAS RNAi lines of selected modifiers mated with either GMR-GAL4 or GMR-GAL4/Cyo;UAS-G4C2-58-GFP/TM6 to induce knockdown of the selected gene. RNA was obtained from whole Drosophila head lysates. Gene expression levels are mean ± s.d. from n = 3 independent experiments, *P < 0.05, **P < 0.01, ***P < 0.001 by one-way ANOVA, Tukey’s post hoc test. g, h, qRT–PCR analysis demonstrating knockdown of Ref1 and Nup50 in salivary gland. mRNA levels of selected modifier (left), GAL4 (middle) and (G4C2)58 (right) assayed by qRT–PCR in progeny resulting from either P(PZ)Ref102267 (g) or Nup5020824/GD (h) mated with Fkh-GAL4,UAS-G4C2-58-GFP/TM6. RNA was obtained from salivary gland lysates. Gene expression levels are mean ± s.d., n = 3 independent experiments. *P < 0.05 by Student’s t-test.

Supplementary information

Supplementary Information

This file contains Supplementary Tables 1-6. (PDF 218 kb)

PowerPoint slides

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Freibaum, B., Lu, Y., Lopez-Gonzalez, R. et al. GGGGCC repeat expansion in C9orf72 compromises nucleocytoplasmic transport. Nature 525, 129–133 (2015). https://doi.org/10.1038/nature14974

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nature14974

This article is cited by

Comments

By submitting a comment you agree to abide by our Terms and Community Guidelines. If you find something abusive or that does not comply with our terms or guidelines please flag it as inappropriate.

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