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:

Haploinsufficiency leads to neurodegeneration in C9ORF72 ALS/FTD human induced motor neurons

Abstract

An intronic GGGGCC repeat expansion in C9ORF72 is the most common cause of amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD), but the pathogenic mechanism of this repeat remains unclear. Using human induced motor neurons (iMNs), we found that repeat-expanded C9ORF72 was haploinsufficient in ALS. We found that C9ORF72 interacted with endosomes and was required for normal vesicle trafficking and lysosomal biogenesis in motor neurons. Repeat expansion reduced C9ORF72 expression, triggering neurodegeneration through two mechanisms: accumulation of glutamate receptors, leading to excitotoxicity, and impaired clearance of neurotoxic dipeptide repeat proteins derived from the repeat expansion. Thus, cooperativity between gain- and loss-of-function mechanisms led to neurodegeneration. Restoring C9ORF72 levels or augmenting its function with constitutively active RAB5 or chemical modulators of RAB5 effectors rescued patient neuron survival and ameliorated neurodegenerative processes in both gain- and loss-of-function C9ORF72 mouse models. Thus, modulating vesicle trafficking was able to rescue neurodegeneration caused by the C9ORF72 repeat expansion. Coupled with rare mutations in ALS2, FIG4, CHMP2B, OPTN and SQSTM1, our results reveal mechanistic convergence on vesicle trafficking in ALS and FTD.

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: C9ORF72 patient iMNs undergo rapid neurodegeneration.
Figure 2: C9ORF72 protein levels determine iMN survival.
Figure 3: Reduced C9ORF72 activity disrupts vesicle trafficking and lysosomal biogenesis in motor neurons.
Figure 4: Low C9ORF72 activity sensitizes neurons to glutamate.
Figure 5: C9ORF72 levels determine dipeptide repeat turnover.
Figure 6: Small molecule modulators of vesicle trafficking rescue neurodegeneration in vitro and in vivo.

Similar content being viewed by others

Accession codes

Accessions

BioProject

Gene Expression Omnibus

References

  1. Renton, A.E., Chiò, A. & Traynor, B.J. State of play in amyotrophic lateral sclerosis genetics. Nat. Neurosci. 17, 17–23 (2014).

    Article  CAS  PubMed  Google Scholar 

  2. Majounie, E. et al. Frequency of the C9orf72 hexanucleotide repeat expansion in patients with amyotrophic lateral sclerosis and frontotemporal dementia: a cross-sectional study. Lancet Neurol. 11, 323–330 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. 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  PubMed  PubMed Central  Google Scholar 

  4. 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  PubMed  PubMed Central  Google Scholar 

  5. Zhang, Y. et al. An RNA-sequencing transcriptome and splicing database of glia, neurons, and vascular cells of the cerebral cortex. J. Neurosci. 34, 11929–11947 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. 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  PubMed  PubMed Central  CAS  Google Scholar 

  7. Donnelly, C.J. et al. RNA toxicity from the ALS/FTD C9ORF72 expansion is mitigated by antisense intervention. Neuron 80, 415–428 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Lagier-Tourenne, C. et al. Targeted degradation of sense and antisense C9orf72 RNA foci as therapy for ALS and frontotemporal degeneration. Proc. Natl. Acad. Sci. USA 110, E4530–E4539 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Haeusler, A.R. et al. C9orf72 nucleotide repeat structures initiate molecular cascades of disease. Nature 507, 195–200 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. 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  PubMed  PubMed Central  Google Scholar 

  11. Mori, K. et al. The C9orf72 GGGGCC repeat is translated into aggregating dipeptide-repeat proteins in FTLD/ALS. Science 339, 1335–1338 (2013).

    Article  CAS  PubMed  Google Scholar 

  12. 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  CAS  PubMed  PubMed Central  Google Scholar 

  13. Su, Z. et al. Discovery of a biomarker and lead small molecules to target r(GGGGCC)-associated defects in c9FTD/ALS. Neuron 84, 239 (2014).

    Article  CAS  PubMed  Google Scholar 

  14. Jiang, J. et al. Gain of toxicity from ALS/FTD-linked repeat expansions in C9ORF72 is alleviated by antisense oligonucleotides targeting GGGGCC-containing RNAs. Neuron 90, 535–550 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Chew, J. et al. Neurodegeneration. C9ORF72 repeat expansions in mice cause TDP-43 pathology, neuronal loss and behavioral deficits. Science 348, 1151–1154 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Liu, Y. et al. C9orf72 BAC mouse model with motor deficits and neurodegenerative features of ALS/FTD. Neuron 90, 521–534 (2016).

    Article  CAS  PubMed  Google Scholar 

  17. Liu, E.Y. et al. C9orf72 hypermethylation protects against repeat expansion-associated pathology in ALS/FTD. Acta Neuropathol. 128, 525–541 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. O'Rourke, J.G. et al. C9orf72 is required for proper macrophage and microglial function in mice. Science 351, 1324–1329 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Burberry, A. et al. Loss-of-function mutations in the C9ORF72 mouse ortholog cause fatal autoimmune disease. Sci. Transl. Med. 8, 347ra93 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  20. Sellier, C. et al. Loss of C9ORF72 impairs autophagy and synergizes with polyQ Ataxin-2 to induce motor neuron dysfunction and cell death. EMBO J. 35, 1276–1297 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Webster, C.P. et al. The C9orf72 protein interacts with Rab1a and the ULK1 complex to regulate initiation of autophagy. EMBO J. 35, 1656–1676 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Koppers, M. et al. C9orf72 ablation in mice does not cause motor neuron degeneration or motor deficits. Ann. Neurol. 78, 426–438 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Fratta, P. et al. Homozygosity for the C9orf72 GGGGCC repeat expansion in frontotemporal dementia. Acta Neuropathol. 126, 401–409 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Son, E.Y. et al. Conversion of mouse and human fibroblasts into functional spinal motor neurons. Cell Stem Cell 9, 205–218 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Marchetto, M.C. et al. Non-cell-autonomous effect of human SOD1 G37R astrocytes on motor neurons derived from human embryonic stem cells. Cell Stem Cell 3, 649–657 (2008).

    Article  CAS  PubMed  Google Scholar 

  26. Kiskinis, E. et al. Pathways disrupted in human ALS motor neurons identified through genetic correction of mutant SOD1. Cell Stem Cell 14, 781–795 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Lin, C.L. et al. Aberrant RNA processing in a neurodegenerative disease: the cause for absent EAAT2, a glutamate transporter, in amyotrophic lateral sclerosis. Neuron 20, 589–602 (1998).

    Article  CAS  PubMed  Google Scholar 

  28. Du, Z.W. et al. Generation and expansion of highly pure motor neuron progenitors from human pluripotent stem cells. Nat. Commun. 6, 6626 (2015).

    Article  CAS  PubMed  Google Scholar 

  29. Pfisterer, U. et al. Direct conversion of human fibroblasts to dopaminergic neurons. Proc. Natl. Acad. Sci. USA 108, 10343–10348 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Aoki, Y. et al. C9orf72 and RAB7L1 regulate vesicle trafficking in amyotrophic lateral sclerosis and frontotemporal dementia. Brain 140, 887–897 (2017).

    Article  PubMed  Google Scholar 

  31. Sullivan, P.M. et al. The ALS/FTLD associated protein C9orf72 associates with SMCR8 and WDR41 to regulate the autophagy-lysosome pathway. Acta Neuropathol. Commun. 4, 51 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  32. Farg, M.A. et al. C9ORF72, implicated in amytrophic lateral sclerosis and frontotemporal dementia, regulates endosomal trafficking. Hum. Mol. Genet. 23, 3579–3595 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Prudencio, M. et al. Distinct brain transcriptome profiles in C9orf72-associated and sporadic ALS. Nat. Neurosci. 18, 1175–1182 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Highley, J.R. et al. Loss of nuclear TDP-43 in amyotrophic lateral sclerosis (ALS) causes altered expression of splicing machinery and widespread dysregulation of RNA splicing in motor neurones. Neuropathol. Appl. Neurobiol. 40, 670–685 (2014).

    Article  CAS  PubMed  Google Scholar 

  35. Levine, T.P., Daniels, R.D., Gatta, A.T., Wong, L.H. & Hayes, M.J. The product of C9orf72, a gene strongly implicated in neurodegeneration, is structurally related to DENN Rab-GEFs. Bioinformatics 29, 499–503 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Marat, A.L., Dokainish, H. & McPherson, P.S. DENN domain proteins: regulators of Rab GTPases. J. Biol. Chem. 286, 13791–13800 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Amick, J., Roczniak-Ferguson, A. & Ferguson, S.M. C9orf72 binds SMCR8, localizes to lysosomes, and regulates mTORC1 signaling. Mol. Biol. Cell 27, 3040–3051 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Neiss, W.F. The electron density of light and dark lysosomes in the proximal convoluted tubule of the rat kidney. Histochemistry 77, 63–77 (1983).

    Article  CAS  PubMed  Google Scholar 

  39. Saftig, P. & Klumperman, J. Lysosome biogenesis and lysosomal membrane proteins: trafficking meets function. Nat. Rev. Mol. Cell Biol. 10, 623–635 (2009).

    Article  CAS  PubMed  Google Scholar 

  40. Ganley, I.G., Carroll, K., Bittova, L. & Pfeffer, S. Rab9 GTPase regulates late endosome size and requires effector interaction for its stability. Mol. Biol. Cell 15, 5420–5430 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Rodriguez-Gabin, A.G., Yin, X., Si, Q. & Larocca, J.N. Transport of mannose-6-phosphate receptors from the trans-Golgi network to endosomes requires Rab31. Exp. Cell Res. 315, 2215–2230 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Waguri, S. et al. Visualization of TGN to endosome trafficking through fluorescently labeled MPR and AP-1 in living cells. Mol. Biol. Cell 14, 142–155 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Wang, G., Gilbert, J. & Man, H.Y. AMPA receptor trafficking in homeostatic synaptic plasticity: functional molecules and signaling cascades. Neural Plast. 2012, 825364 (2012).

    PubMed  PubMed Central  Google Scholar 

  44. Chen, P., Gu, Z., Liu, W. & Yan, Z. Glycogen synthase kinase 3 regulates N-methyl-D-aspartate receptor channel trafficking and function in cortical neurons. Mol. Pharmacol. 72, 40–51 (2007).

    Article  CAS  PubMed  Google Scholar 

  45. Chen, T.W. et al. Ultrasensitive fluorescent proteins for imaging neuronal activity. Nature 499, 295–300 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Wainger, B.J. et al. Intrinsic membrane hyperexcitability of amyotrophic lateral sclerosis patient-derived motor neurons. Cell Rep. 7, 1–11 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Gurskaya, N.G. et al. Engineering of a monomeric green-to-red photoactivatable fluorescent protein induced by blue light. Nat. Biotechnol. 24, 461–465 (2006).

    Article  CAS  PubMed  Google Scholar 

  48. Mackenzie, I.R. et al. Quantitative analysis and clinico-pathological correlations of different dipeptide repeat protein pathologies in C9ORF72 mutation carriers. Acta Neuropathol. 130, 845–861 (2015).

    Article  CAS  PubMed  Google Scholar 

  49. Cai, X. et al. PIKfyve, a class III PI kinase, is the target of the small molecular IL-12/IL-23 inhibitor apilimod and a player in Toll-like receptor signaling. Chem. Biol. 20, 912–921 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Lemmon, M.A. Membrane recognition by phospholipid-binding domains. Nat. Rev. Mol. Cell Biol. 9, 99–111 (2008).

    Article  CAS  PubMed  Google Scholar 

  51. Martin, S. et al. Inhibition of PIKfyve by YM-201636 dysregulates autophagy and leads to apoptosis-independent neuronal cell death. PLoS One 8, e60152 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Ikonomov, O.C., Sbrissa, D. & Shisheva, A. Localized PtdIns 3,5-P2 synthesis to regulate early endosome dynamics and fusion. Am. J. Physiol. Cell Physiol. 291, C393–C404 (2006).

    Article  CAS  PubMed  Google Scholar 

  53. Chow, C.Y. et al. Deleterious variants of FIG4, a phosphoinositide phosphatase, in patients with ALS. Am. J. Hum. Genet. 84, 85–88 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Lodhi, I.J. et al. Insulin stimulates phosphatidylinositol 3-phosphate production via the activation of Rab5. Mol. Biol. Cell 19, 2718–2728 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Zhong, Z. et al. Protein S protects neurons from excitotoxic injury by activating the TAM receptor Tyro3-phosphatidylinositol 3-kinase-Akt pathway through its sex hormone-binding globulin-like region. J. Neurosci. 30, 15521–15534 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. O'Rourke, J.G. et al. C9orf72 BAC transgenic mice display typical pathologic features of ALS/FTD. Neuron 88, 892–901 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Zeigerer, A. et al. Rab5 is necessary for the biogenesis of the endolysosomal system in vivo. Nature 485, 465–470 (2012).

    Article  CAS  PubMed  Google Scholar 

  58. Topp, J.D., Gray, N.W., Gerard, R.D. & Horazdovsky, B.F. Alsin is a Rab5 and Rac1 guanine nucleotide exchange factor. J. Biol. Chem. 279, 24612–24623 (2004).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  60. Ran, F.A. et al. Genome engineering using the CRISPR-Cas9 system. Nat. Protoc. 8, 2281–2308 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Li, J. et al. Long-term potentiation modulates synaptic phosphorylation networks and reshapes the structure of the postsynaptic interactome. Sci. Signal. 9, rs8 (2016).

    Article  CAS  PubMed  Google Scholar 

  62. Park, I.H. et al. Reprogramming of human somatic cells to pluripotency with defined factors. Nature 451, 141–146 (2008).

    Article  CAS  PubMed  Google Scholar 

  63. Dobin, A. et al. STAR: ultrafast universal RNA-seq aligner. Bioinformatics 29, 15–21 (2013).

    Article  CAS  PubMed  Google Scholar 

  64. Anders, S. et al. Count-based differential expression analysis of RNA sequencing data using R and Bioconductor. Nat. Protoc. 8, 1765–1786 (2013).

    Article  CAS  PubMed  Google Scholar 

  65. Yu, G., Wang, L.-G., Han, Y. & He, Q.-Y. clusterProfiler: an R package for comparing biological themes among gene clusters. OMICS 16, 284–287 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank the NINDS Biorepository at Coriell Institute for providing the following cell lines for this study: ND12133, ND03231, ND01751, ND11976, ND03719, ND00184, ND5280, ND06769, ND10689, ND12099, ND14954, ND08957, ND12100 and ND014587. We thank H. Chui and C. Miller (University of Southern California Alzheimer's Disease Research Center) and N. Shneider (Columbia University Medical Center) for control and C9ORF72 patient tissue. We thank the Choi Family Therapeutic Screening Facility for chemical screening support and the Translational Imaging Center at USC for imaging support. We thank M. Koppers, Y, Adolfs, C. van der Meer and M. Broekhoven for help with mouse breeding and kainate injection experiments. We thank S. Waguri (Fukushima Medical University) for providing the M6PR-GFP construct. We thank C, Buser for assistance with electron microscopy. We also thank S. Alworth (DRVision Technologies), K. Hebestreit and R. Bhatnagar (Verge Genomics), B. Baloh (Cedars Sinai Medical Center), J. O'Rourke (Cedars Sinai Medical Center), C. Donnelly, C. Tong, A. McMahon and Q. Liu-Michael for reagents, technical support and discussions. E.Y.S. is a Walter V. and Idun Berry Postdoctoral Fellow. K.A.S. was supported in part by a Muscular Dystrophy Association Development Grant. L.M. was supported by NIH grant T32DC009975-04. This work was supported by NIH grants AG039452, AG023084 and NS034467 to B.V.Z. R.J.P. was supported by grants from ALS Foundation Netherlands (TOTALS), Epilepsiefonds (12-08, 15-05), and VICI grant Netherlands Organization for Scientific Research (NWO). This work was also supported by NIH grants R00NS077435 and R01NS097850, US Department of Defense grant W81XWH-15-1-0187, and grants from the Donald E. and Delia B. Baxter Foundation, the Tau Consortium, the Frick Foundation for ALS Research, the Muscular Dystrophy Association, the New York Stem Cell Foundation, the USC Keck School of Medicine Regenerative Medicine Initiative, the USC Broad Innovation Award, and the Southern California Clinical and Translational Science Institute to J.K.I. J.K.I. is a New York Stem Cell Foundation-Robertson Investigator.

Author information

Authors and Affiliations

Authors

Contributions

Y.S., S.L., Y.L. and J.K.I. conceived the project. Y.S., S.L., E.Y.S., Y.L., L.M., K.A.S., V.R.V., K.S., S.J.J.L., P.R.A., M.C., R.J.P., D.T., B.V.Z. and J.K.I. designed the experiments. Y.S., S.L., W.-H.C., E.Y.S., Y.L., S.-T.H., E.H., G.R.L., T.S., M.H., C.S., A.R.N., T.-Y.C., Y.W., K.K., B.W., L.M., M.J.C., B.G., K.P.S., J. K., N.K., X.W., V.H., A.R.N., K.A.S., V.R.V., K.S., R.J.P. and J.K.I. performed experiments and interpreted data. K.K. performed all electrophysiological studies and P.W., J.A.C., N.H.-S., N.W., T.G.B., A.Z. and K.A.S. performed RNA-Seq analysis. Y.S., S.L., E.Y.S., K.A.S. and J.K.I. prepared the manuscript. C.G. and M.W. developed the method of inducing iMNs using the Dox-NIL construct. All of the authors discussed the results and commented on the manuscript.

Corresponding author

Correspondence to Justin K Ichida.

Ethics declarations

Competing interests

J.K.I. and P.A. are co-founders of Acurastem, Inc. P.A. is an employee of Icagen Corporation. J.K.I. and P.A. declare that they are bound by confidentiality agreements that prevent them from disclosing details of their financial interests in this work. S.-J.L. is a founder of DRVision Technologies and T.-Y.C. is an employee of DRVision Technologies. A.Z. and J.A.C. are co-founders of Verge Genomics and A.Z., V.H.-S., N.W. and T.G.B. are employees of Verge Genomics.

Supplementary information

Supplementary Figures & Tables

Supplementary Figures 1–17 & Supplementary Tables 1–6 (PDF 39872 kb)

Life Sciences Reporting Summary (PDF 247 kb)

Supplementary Table 7

RNA sequencing data of Hb9::RFP+ iMNs from CTRL2, C9- ALS1, CTRL2 C9ORF72+/-, and CTRL2 C9ORF72-/- iPSCs. (XLSX 11015 kb)

41591_2018_BFnm4490_MOESM4_ESM.mov

Channel rhodopsin neuromuscular junction assay: C9-ALS patient iMNs (C9-ALS2). Green light is activated and can be observed by an increase in overall brightness at the following times: 7-13 sec, 18-23 sec, 29-35 sec, 40-45 sec, 51-56 sec, 1:02-1:07. Light-induced myotube contraction can be observed during those intervals. (MOV 11016 kb)

41591_2018_BFnm4490_MOESM5_ESM.mov

Channel rhodopsin neuromuscular junction assay: control iMNs (CTRL2). Green light is activated and can be observed by an increase in overall brightness at the following times: 7- 12 sec, 18-23 sec, 29-34 sec, 40-45 sec, 52-57 sec, 1:03-1:08. Light-induced myotube contraction can be observed during those intervals. (MOV 16834 kb)

41591_2018_BFnm4490_MOESM6_ESM.mov

Time-lapse video of C9-ALS (C9-ALS3) iMN degeneration. Fluorescent neurons are Hb9::RFP+ iMNs. Frames were captured at 24-hour intervals. (MOV 9143 kb)

41591_2018_BFnm4490_MOESM7_ESM.mov

Time-lapse video of control (CTRL2) iMN degeneration. Fluorescent neurons are Hb9::RFP+ iMNs. Frames were captured at 24-hour intervals. (MOV 10366 kb)

41591_2018_BFnm4490_MOESM8_ESM.mov

Time-lapse video of M6PR-GFP+ vesicle trafficking in a control iMN. Frames were captured at a rate of 8 frames/sec over a 60 sec interval. The green line outlines the cell soma. (MOV 628 kb)

41591_2018_BFnm4490_MOESM9_ESM.mov

Time-lapse video of M6PR-GFP+ vesicle trafficking in a C9ORF72+/- (CTRL2, C9+/-) iMN. Frames were captured at a rate of 8 frames/sec over a 60 sec interval. The white line outlines the cell soma. (MOV 230 kb)

41591_2018_BFnm4490_MOESM10_ESM.mov

Time-lapse video of M6PR-GFP+ vesicle trafficking in a C9- ALS iMN. Frames were captured at a rate of 8 frames/sec over a 60 sec interval. The white line outlines the cell soma. (MOV 216 kb)

41591_2018_BFnm4490_MOESM11_ESM.mov

Time-lapse video of M6PR-GFP+ vesicle trafficking in a C9- ALS iMN expressing C9ORF72 isoform A. Frames were captured at a rate of 8 frames/sec over a 60 sec interval. The white line outlines the cell soma. (MOV 486 kb)

41591_2018_BFnm4490_MOESM12_ESM.mov

Time-lapse video of M6PR-GFP+ vesicle trafficking in a C9- ALS iMN expressing C9ORF72 isoform B. Frames were captured at a rate of 8 frames/sec over a 60 sec interval. The white line outlines the cell soma. (MOV 487 kb)

41591_2018_BFnm4490_MOESM13_ESM.mov

Time-lapse video of Gcamp6 fluorescence in control iMNs treated with glutamate. Frames were captured over a 24 second interval and the video was increased to 2x speed to facilitate viewing. (MOV 139 kb)

41591_2018_BFnm4490_MOESM14_ESM.mov

Time-lapse video of Gcamp6 fluorescence in C9-ALS iMNs treated with glutamate. Frames were captured over a 24 second interval and the video was increased to 2x speed to facilitate viewing. (MOV 233 kb)

41591_2018_BFnm4490_MOESM15_ESM.mov

Time-lapse video of Gcamp6 fluorescence in C9ORF72+/- iMNs treated with glutamate. Frames were captured over a 24 second interval and the video was increased to 2x speed to facilitate viewing. (MOV 156 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Shi, Y., Lin, S., Staats, K. et al. Haploinsufficiency leads to neurodegeneration in C9ORF72 ALS/FTD human induced motor neurons. Nat Med 24, 313–325 (2018). https://doi.org/10.1038/nm.4490

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nm.4490

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