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:

Globally reduced N6-methyladenosine (m6A) in C9ORF72-ALS/FTD dysregulates RNA metabolism and contributes to neurodegeneration

Abstract

Repeat expansion in C9ORF72 is the most common genetic cause of amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD). Here we show that N6-methyladenosine (m6A), the most prevalent internal mRNA modification, is downregulated in C9ORF72-ALS/FTD patient-derived induced pluripotent stem cell (iPSC)-differentiated neurons and postmortem brain tissues. The global m6A hypomethylation leads to transcriptome-wide mRNA stabilization and upregulated gene expression, particularly for genes involved in synaptic activity and neuronal function. Moreover, the m6A modification in the C9ORF72 intron sequence upstream of the expanded repeats enhances RNA decay via the nuclear reader YTHDC1, and the antisense RNA repeats can also be regulated through m6A modification. The m6A reduction increases the accumulation of repeat RNAs and the encoded poly-dipeptides, contributing to disease pathogenesis. We further demonstrate that, by elevating m6A methylation, we could significantly reduce repeat RNA levels from both strands and the derived poly-dipeptides, rescue global mRNA homeostasis and improve survival of C9ORF72-ALS/FTD patient iPSC-derived neurons.

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

Fig. 1: The expression of m6A methyltransferases and the m6A RNA modification levels are downregulated in C9ORF72-ALS/FTD.
Fig. 2: m6A hypomethylation dysregulates neuronal gene expression in C9ORF72-ALS/FTD.
Fig. 3: m6A hypomethylation increases mRNA half-life via YTHDF2 in iPSNs.
Fig. 4: The m6A modification in the repeat-containing intron modulates DPR protein production.
Fig. 5: YTHDC1 regulates the stability of C9ORF72 repeat-containing intron RNA via the NEXT decay pathway.
Fig. 6: m6A restoration rescues disease-related phenotypes in C9ORF72-ALS/FTD patient iPSNs.
Fig. 7: FTO inhibitor treatment mitigates disease phenotypes.

Similar content being viewed by others

Data availability

Requests for further information or resources and reagents should be directed to and will be filled by the lead author, Shuying Sun (shuying.sun@jhmi.edu). Plasmids generated in this study are available from the lead author upon reasonable request. The sequencing data (Supplementary Table 7) have been deposited at the National Center for Biotechnology Information under Gene Expression Omnibus accession number GSE203581. Source data are provided with this paper.

Code availability

This paper does not report original code.

References

  1. Nussbacher, J. K., Tabet, R., Yeo, G. W. & Lagier-Tourenne, C. Disruption of RNA metabolism in neurological diseases and emerging therapeutic interventions. Neuron 102, 294–320 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  2. Ferrari, R., Kapogiannis, D., Huey, E. D. & Momeni, P. FTD and ALS: a tale of two diseases. Curr. Alzheimer Res. 8, 273–294 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  3. Ito, D., Hatano, M. & Suzuki, N. RNA binding proteins and the pathological cascade in ALS/FTD neurodegeneration. Sci. Transl. Med. 9, eaah5436 (2017).

    PubMed  Google Scholar 

  4. Freibaum, B. D. & Taylor, J. P. The role of dipeptide repeats in C9ORF72-related ALS-FTD. Front. Mol. Neurosci. 10, 35 (2017).

    PubMed  PubMed Central  Google Scholar 

  5. Cheng, W. et al. CRISPR–Cas9 screens identify the RNA helicase DDX3X as a repressor of C9ORF72 (GGGGCC)n repeat-associated non-AUG translation. Neuron 104, 885–898 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  6. Dominissini, D. et al. Topology of the human and mouse m6A RNA methylomes revealed by m6A-seq. Nature 485, 201–206 (2012).

    CAS  PubMed  Google Scholar 

  7. Meyer, K. D. et al. Comprehensive analysis of mRNA methylation reveals enrichment in 3′ UTRs and near stop codons. Cell 149, 1635–1646 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  8. Liu, J. et al. A METTL3–METTL14 complex mediates mammalian nuclear RNA N6-adenosine methylation. Nat. Chem. Biol. 10, 93–95 (2014).

    CAS  PubMed  Google Scholar 

  9. Roundtree, I. A., Evans, M. E., Pan, T. & He, C. Dynamic RNA modifications in gene expression regulation. Cell 169, 1187–1200 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  10. Jia, G. et al. N6-methyladenosine in nuclear RNA is a major substrate of the obesity-associated FTO. Nat. Chem. Biol. 7, 885–887 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  11. Zheng, G. et al. ALKBH5 is a mammalian RNA demethylase that impacts RNA metabolism and mouse fertility. Mol. Cell 49, 18–29 (2013).

    CAS  PubMed  Google Scholar 

  12. Xiao, W. et al. Nuclear m6A reader YTHDC1 regulates mRNA splicing. Mol. Cell 61, 507–519 (2016).

    CAS  PubMed  Google Scholar 

  13. Du, H. et al. YTHDF2 destabilizes m6A-containing RNA through direct recruitment of the CCR4–NOT deadenylase complex. Nat. Commun. 7, 12626 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  14. Wang, X. et al. N6-methyladenosine modulates messenger RNA translation efficiency. Cell 161, 1388–1399 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  15. Wang, X. et al. N6-methyladenosine-dependent regulation of messenger RNA stability. Nature 505, 117–120 (2014).

    PubMed  Google Scholar 

  16. Liu, J. et al. Landscape and regulation of m6A and m6Am methylome across human and mouse tissues. Mol. Cell 77, 426–440 (2020).

    CAS  PubMed  Google Scholar 

  17. Livneh, I., Moshitch-Moshkovitz, S., Amariglio, N., Rechavi, G. & Dominissini, D. The m6A epitranscriptome: transcriptome plasticity in brain development and function. Nat. Rev. Neurosci. 21, 36–51 (2020).

    CAS  PubMed  Google Scholar 

  18. Deng, X. et al. Widespread occurrence of N6-methyladenosine in bacterial mRNA. Nucleic Acids Res. 43, 6557–6567 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  19. Tian, R. et al. CRISPR interference-based platform for multimodal genetic screens in human iPSC-derived neurons. Neuron 104, 239–255 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  20. 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).

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  Google Scholar 

  22. Cheng, W. et al. C9ORF72 GGGGCC repeat-associated non-AUG translation is upregulated by stress through eIF2α phosphorylation. Nat. Commun. 9, 51 (2018).

    PubMed  PubMed Central  Google Scholar 

  23. Wang, S. et al. Nuclear export and translation of circular repeat-containing intronic RNA in C9ORF72-ALS/FTD. Nat. Commun. 12, 4908 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  24. Liu, J. et al. N6-methyladenosine of chromosome-associated regulatory RNA regulates chromatin state and transcription. Science 367, 580–586 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  25. Lubas, M. et al. Interaction profiling identifies the human nuclear exosome targeting complex. Mol. Cell 43, 624–637 (2011).

    CAS  PubMed  Google Scholar 

  26. Mori, K. et al. Bidirectional transcripts of the expanded C9orf72 hexanucleotide repeat are translated into aggregating dipeptide repeat proteins. Acta Neuropathol. 126, 881–893 (2013).

    CAS  PubMed  Google Scholar 

  27. 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).

    CAS  PubMed  PubMed Central  Google Scholar 

  28. 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).

    CAS  PubMed  PubMed Central  Google Scholar 

  29. Mizielinska, S. et al. C9orf72 frontotemporal lobar degeneration is characterised by frequent neuronal sense and antisense RNA foci. Acta Neuropathol. 126, 845–857 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  30. Zu, T. et al. RAN proteins and RNA foci from antisense transcripts in C9ORF72 ALS and frontotemporal dementia. Proc. Natl Acad. Sci. USA 110, E4968–E4977 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  31. 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).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  33. Huang, Y. et al. Small-molecule targeting of oncogenic FTO demethylase in acute myeloid leukemia. Cancer Cell 35, 677–691.e10 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  34. Su, R. et al. Targeting FTO suppresses cancer stem cell maintenance and immune evasion. Cancer Cell 38, 79–96 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  35. Liu, Y. et al. Tumors exploit FTO-mediated regulation of glycolytic metabolism to evade immune surveillance. Cell Metab. 33, 1221–1233 (2021).

    CAS  PubMed  Google Scholar 

  36. Jiang, X. et al. The role of m6A modification in the biological functions and diseases. Signal Transduct. Target Ther. 6, 74 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  37. Lence, T. et al. m6A modulates neuronal functions and sex determination in Drosophila. Nature 540, 242–247 (2016).

    CAS  PubMed  Google Scholar 

  38. Wang, C. X. et al. METTL3-mediated m6A modification is required for cerebellar development. PLoS Biol. 16, e2004880 (2018).

    PubMed  PubMed Central  Google Scholar 

  39. Devlin, A. C. et al. Human iPSC-derived motoneurons harbouring TARDBP or C9ORF72 ALS mutations are dysfunctional despite maintaining viability. Nat. Commun. 6, 5999 (2015).

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  41. Koranda, J. L. et al. Mettl14 is essential for epitranscriptomic regulation of striatal function and learning. Neuron 99, 283–292 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  42. Lall, D. et al. C9orf72 deficiency promotes microglial-mediated synaptic loss in aging and amyloid accumulation. Neuron 109, 2275–2291 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  43. Zhu, Q. et al. Reduced C9ORF72 function exacerbates gain of toxicity from ALS/FTD-causing repeat expansion in C9orf72. Nat. Neurosci. 23, 615–624 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  44. Schmitz, A., Pinheiro Marques, J., Oertig, I., Maharjan, N. & Saxena, S. Emerging perspectives on dipeptide repeat proteins in C9ORF72 ALS/FTD. Front. Cell. Neurosci. 15, 637548 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  45. McEachin, Z. T., Parameswaran, J., Raj, N., Bassell, G. J. & Jiang, J. RNA-mediated toxicity in C9orf72 ALS and FTD. Neurobiol. Dis. 145, 105055 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  46. Donnelly, C. J., Grima, J. C. & Sattler, R. Aberrant RNA homeostasis in amyotrophic lateral sclerosis: potential for new therapeutic targets? Neurodegener. Dis. Manag. 4, 417–437 (2014).

    PubMed  Google Scholar 

  47. Conlon, E. G. et al. The C9ORF72 GGGGCC expansion forms RNA G-quadruplex inclusions and sequesters hnRNP H to disrupt splicing in ALS brains. eLife 5, e17820 (2016).

    PubMed  PubMed Central  Google Scholar 

  48. Lee, Y. B. et al. Hexanucleotide repeats in ALS/FTD form length-dependent RNA foci, sequester RNA binding proteins, and are neurotoxic. Cell Rep. 5, 1178–1186 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  49. 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).

    PubMed  PubMed Central  Google Scholar 

  50. 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).

    CAS  PubMed  PubMed Central  Google Scholar 

  51. McMillan, M. et al. RNA methylation influences TDP43 binding and disease pathogenesis in models of amyotrophic lateral sclerosis and frontotemporal dementia. Mol. Cell. 83, 219–236 (2023).

    CAS  PubMed  Google Scholar 

  52. Zhao, F. et al. METTL3-dependent RNA m6A dysregulation contributes to neurodegeneration in Alzheimer’s disease through aberrant cell cycle events. Mol. Neurodegener. 16, 70 (2021).

    PubMed  PubMed Central  Google Scholar 

  53. Alzheimer’s Association. 2016 Alzheimer’s disease facts and figures. Alzheimers Dement. 12, 459–509 (2016).

  54. Shafik, A. M. et al. N6-methyladenosine dynamics in neurodevelopment and aging, and its potential role in Alzheimer’s disease. Genome Biol. 22, 17 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  55. Chen, X. et al. Down-regulation of m6A mRNA methylation is involved in dopaminergic neuronal death. ACS Chem. Neurosci. 10, 2355–2363 (2019).

    CAS  PubMed  Google Scholar 

  56. Ababneh, N. A. et al. Correction of amyotrophic lateral sclerosis related phenotypes in induced pluripotent stem cell-derived motor neurons carrying a hexanucleotide expansion mutation in C9orf72 by CRISPR/Cas9 genome editing using homology-directed repair. Hum. Mol. Genet. 29, 2200–2217 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  57. Cleary, E. M. et al. Improved PCR based methods for detecting C9orf72 hexanucleotide repeat expansions. Mol. Cell Probes 30, 218–224 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  58. Zaepfel, B. L. et al. UPF1 reduces C9orf72 HRE-induced neurotoxicity in the absence of nonsense-mediated decay dysfunction. Cell Rep. 34, 108925 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  59. Coyne, A. N. et al. Nuclear accumulation of CHMP7 initiates nuclear pore complex injury and subsequent TDP-43 dysfunction in sporadic and familial ALS. Sci. Transl. Med. 13, eabe1923 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  60. Bolger, A. M., Lohse, M. & Usadel, B. Trimmomatic: a flexible trimmer for Illumina sequence data. Bioinformatics 30, 2114–2120 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  61. Kim, D., Paggi, J. M., Park, C., Bennett, C. & Salzberg, S. L. Graph-based genome alignment and genotyping with HISAT2 and HISAT-genotype. Nat. Biotechnol. 37, 907–915 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  62. Danecek, P. et al. Twelve years of SAMtools and BCFtools. Gigascience 10, giab008 (2021).

    PubMed  PubMed Central  Google Scholar 

  63. Zhang, Y. et al. Model-based analysis of ChIP-seq (MACS). Genome Biol. 9, R137 (2008).

    PubMed  PubMed Central  Google Scholar 

  64. Cui, X. et al. MeTDiff: a novel differential RNA methylation analysis for MeRIP-seq data. IEEE/ACM Trans. Comput. Biol. Bioinform. 15, 526–534 (2018).

    CAS  PubMed  Google Scholar 

  65. Anders, S., Pyl, P. T. & Huber, W. HTSeq—a Python framework to work with high-throughput sequencing data. Bioinformatics 31, 166–169 (2015).

    CAS  PubMed  Google Scholar 

  66. Love, M. I., Huber, W. & Anders, S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol. 15, 550 (2014).

    PubMed  PubMed Central  Google Scholar 

  67. Robinson, M. D., McCarthy, D. J. & Smyth, G. K. edgeR: a Bioconductor package for differential expression analysis of digital gene expression data. Bioinformatics 26, 139–140 (2010).

    CAS  PubMed  Google Scholar 

  68. Heinz, S. et al. Simple combinations of lineage-determining transcription factors prime cis-regulatory elements required for macrophage and B cell identities. Mol. Cell 38, 576–589 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  69. Dennis, G. et al. DAVID: database for annotation, visualization, and integrated discovery. Genome Biol. 4, P3 (2003).

    PubMed  Google Scholar 

  70. Sherman, B. T. et al. DAVID: a web server for functional enrichment analysis and functional annotation of gene lists (2021 update). Nucleic Acids Res. 50, W216–W221 (2022).

    CAS  PubMed  PubMed Central  Google Scholar 

  71. Huang, D. W. et al. DAVID bioinformatics resources: expanded annotation database and novel algorithms to better extract biology from large gene lists. Nucleic Acids Res. 35, W169–W175 (2007).

    PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank Target ALS Postmortem Tissue Core and Johns Hopkins ALS Postmortem Tissue Core for providing postmortem human brain tissue samples. We thank T. Thompson from Answer ALS consortium for providing help and detailed data information. We thank M. Ward and his laboratory at the National Institutes of Health (NIH) for sharing the i3Neuron iPSC line and the i3Neuron differentiation protocols. We thank K. Talbot at the University of Oxford for providing the isogenic pair of patient iPSC lines. We thank L. Xue for help with the GP ELISA. We thank J. Rothstein and his laboratory for sharing control and patient iPSC lines and protocols. We thank Sun laboratory members for helpful discussions. This work is supported by the Johns Hopkins University Catalyst Award (S.S.); NIH grants R01NS107347 (S.S.), RF1NS113820 (S.S.), R21AG072078 (S.S.), RF1NS127925 (S.S.), HG008935 (C.H.), ES030546 (C.H.), K08NS104273 (L.H.) and R01NS123538 (L.H.); and the Packard Center for ALS Research (S.S.). Z.Z. was a recipient of the Milton Safenowitz Post-Doctoral Fellowship from the ALS Association, the Toffler Scholar Award and the Postdoc Development Grant from the Muscular Dystrophy Association. The Mass Spectrometry Facility of the University of Chicago is funded by the National Science Foundation (CHE-1048528). C.H. is an investigator of the Howard Hughes Medical Institute.

Author information

Authors and Affiliations

Authors

Contributions

Y.L., X.D., C.H. and S.S. contributed to the overall design of the study and wrote the paper. Y.L. performed most molecular and cellular biology experiments, with help from Y.X., J.L., Z.Z., R.W., X.F. and Y.Y., under the mentorship of S.S. and C.H. X.D. and Y.L. performed bioinformatic analysis of the high-throughput sequencing data. B.Y. performed blinded analysis of glutamate excitotoxicity. L.H. performed GP ELISA assay and analysis. L.W.O. advised on patient samples and provided key reagents.

Corresponding authors

Correspondence to Chuan He or Shuying Sun.

Ethics declarations

Competing interests

C.H. is a scientific founder, a member of the scientific advisory board and an equity holder of Aferna Bio, Inc. and AccuaDX Inc.; a scientific co-founder and equity holder of Accent Therapeutics, Inc.; and a member of the scientific advisory board of Rona Therapeutics. All other authors declare no competing interests.

Peer review

Peer review information

Nature Neuroscience thanks Adrian Isaacs, Yongchao Ma and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

Additional information

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Extended data

Extended Data Fig. 1 m6A pathway-related gene expression in C9ORF72-ALS/FTD iPSNs.

a, Proteomic heatmap of the m6A pathway components in control (n = 26) and C9ORF72-ALS/FTD (n = 22) iPSNs. Data were normalized to the batch controls. b, Boxplots of the proteomic data in (a). Box plots indicate the interquartile range with the central line representing the median, and the vertical lines extend to the extreme values in the group. c, Western blotting (left) and quantification (right) of METTL3 and METTL14 expression in control and C9ORF72 iPSNs. Data are mean ± s.e.m. ***P = 0.0008 and 0.0003, respectively, by two-tailed t test. d, Relative RNA expression of neuronal marker genes (MAP2, PSD95 and TUBB3) was measured by RT-qPCR in control and C9 iPSNs, showing comparable neuron maturity between control and C9. Points represent individual control or patient lines. n = 6 in each group. Data are mean ± s.e.m. MAP2, P = 0.304; PSD95, P = 0.86; TUBB3, P = 0.716, by two-tailed t test.

Source data

Extended Data Fig. 2 METTL3 and METTL14 m6A methyltransferases were reduced in C9ORF72-ALS/FTD postmortem brain.

a, b, Immunohistochemistry (IHC) staining of METTL3 (a), METTL14 (b) at temporal cortex section of control and C9ORF72-ALS/FTD samples. Scale bar = 50 μm. c, Western blotting (top) and quantification (bottom) of METTL3, METTL14 using postmortem motor cortex tissue samples. *Non-specific bands. Points represent individual control or patient samples. n = 3 in each group. Data are mean ± s.e.m. *P = 0.0171, **P = 0.0011, by two-tailed t test.

Source data

Extended Data Fig. 3 Expression changes of the m6A ‘writer’ core components in sporadic ALS iPSNs.

a, Proteomic heatmap of the core components of m6A ‘writer’ complex in control (n = 19) and sporadic ALS (n = 93) iPSNs. Data were normalized to the batch controls. b, Boxplots of the proteomic data in (a). Box plots indicate the interquartile range with the central line representing the median, and the vertical lines extend to the extreme values in the group. c, UHPLC-QQQ-MS/MS quantification of the m6A/A ratio in poly-A mRNAs from control (n = 5) and sporadic ALS patient (n = 7) postmortem frontal cortex. Data are mean ± s.e.m. Points represent individual samples. *P = 0.0247 by two-tailed t test.

Source data

Extended Data Fig. 4 The repeat RNA induces downregulation of METTL3 and METTL14 expression.

a, Timeline for doxycycline-induced neuron differentiation, lentiviral transduction of transgenes to express repeats or DPRs, and functional analysis of i3Neurons. b, Relative RNA expression of METTL3 and METTL14 in i3Neurons expressing GFP control, modified poly-GA, modified poly-GR (with randomized codons), or (GGGGCC)160 repeats. METTL3: mGA, P = 0.1154; mGR, P = 0.9755; GC160, ****P < 0.0001. METTL14: mGA, P = 0.3892; mGR, P = 0.9853; GC160, **P = 0.0073. P values were calculated by one-way analysis of variance (ANOVA) with Tukey’s multiple comparisons c, Detection of GFP, modified poly-GA and modified poly-GR post lentiviral transduction in i3Neurons by Nano-Glo HiBiT bioluminescence measurement. The luciferase signal was normalized to the total protein level. i3Neurons without lentiviral infection served as the negative control. *P = 0.0148, **P = 0.0062, ***P = 0.0004, by two-tailed t test. d, Immunofluorescent staining of GFP, modified poly-GA or modified poly-GR in transduced i3Neurons. Neurons were stained using FLAG antibody against the C-terminal FLAG epitope tag in each construct. (b,c) Points represent three biological replicates. Data are mean ± s.e.m.

Source data

Extended Data Fig. 5 MeRIP-seq in control and C9ORF72-ALS/FTD iPSNs and postmortem motor cortex.

a, Venn diagram depicting the total number and overlaps of m6A peaks detected in control and C9ORF72-ALS/FTD groups of iPSN (top) and motor cortex (bottom). b, The most enriched consensus m6A motifs in all conditions. c, Metagene profiles of m6A peak density along transcripts with three non-overlapping segments (5’ UTR, CDS, and 3’ UTR) for control and C9ORF72-ALS/FTD groups. d, Principal component analysis (PCA) of control and C9ORF72-ALS/FTD samples. e, Gene Ontology (GO) enrichment analysis of hypomethylated genes that were identified in both iPSNs and motor cortex. P value was calculated by one-sided Fisher’s Exact test.

Extended Data Fig. 6 The impact of m6A hypomethylation on mRNAs.

a, b, Volcano plot of differentially expressed genes in C9ORF72-ALS/FTD compared to the control, in iPSNs (a) and postmortem motor cortex (b). Genes with significant changes are represented by orange (upregulated in C9) and blue (downregulated in C9) dots. iPSNs: n = 4 individual lines per group, 1212 upregulated vs 1168 downregulated. Motor cortex: n = 3 individual tissue samples per group, 1892 upregulated versus 1239 downregulated. P < 0.05, two-tailed Wald test. c, YTHDF2-RIP qPCR of selected YTHDF2 mRNA targets identified from the non-YTHDF2 bound RNAs. HPRT1 and SMARCA1 mRNAs have no m6A modification according to the MeRIP-seq results. Points represent individual iPSN lines. n = 3 in each group. Data are mean ± s.e.m. P = 0.7047 and 0.4001, respectively, by two-tailed t test. d, Cumulative distribution demonstrating the half-life of YTHDF2 targets between control and C9ORF72-ALS/FTD iPSNs. P value was calculated by a two-tailed non-parametric Wilcoxon-Matt-Whitney test. e, Gene Ontology (GO) enrichment analysis of hypomethylated YTHDF2 binding targets which also showed significantly increased half-life. P value was calculated by one-sided Fisher’s Exact test.

Source data

Extended Data Fig. 7 m6A regulates C9ORF72 repeat-containing RNA.

a, IGV profile showing the m6A peaks (generated by comparing MeRIP against input) in control and C9ORF72-ALS/FTD iPSNs. Red arrow head represents the repeat expansion. b, Western blotting of METTL14 in control or METTL14 knockdown i3Neurons. *non-specific band. c, The relative basal expression of NLuc RNA in the HeLa Flp-In cells expressing the reporters as described in Fig. 4b. *P = 0.015 by two-tailed t test. d, The relative expression level of C9R-NLuc RNA upon knockdown of METTL14 in i3Neurons, measured by RT-qPCR and normalized to non-targeting shRNA control. ****P < 0.0001 by two-tailed t test. (c,d) Points represent three biological replicates. Data are mean ± s.e.m.

Source data

Extended Data Fig. 8 The turnover of C9ORF72 repeat-containing RNA is regulated by m6A in i3Neurons.

a, RT-qPCR of C9 repeat RNA expression from the C9R-NLuc HeLa reporter upon knockdown of YTHDF2. P = 0.5418 by two-tailed t test. b, Relative DPR level was measured by luciferase assay upon YTHDF2 knockdown in the C9R-NLuc HeLa reporter. c, Western blotting of YTHDF2 in control or YTHDF2 knockdown HeLa Flp-In cells. P = 0.9456, 0.8801 and 0.9731, respectively, by one-way analysis of variance (ANOVA) with Tukey’s multiple comparisons. d, e, The relative expression level of YTHDC1 RNA (d) or C9R-NLuc RNA (e) upon knockdown of YTHDC1 by shRNA in i3Neurons. **P = 0.0046, ***P = 0.0006 by two-tailed t test. f, g, Relative DPR levels were measured by luciferase assay upon YTHDC1 knockdown (f) or ZCCHC8 knockdown (g) in the i3Neurons expressing the reporters in Fig. 4b. NLuc signal was normalized to the total protein level in each sample and the relative expression was compared to non-targeting shRNA control. ****P < 0.0001 by one-way analysis of variance (ANOVA) with Tukey’s multiple comparisons. h, Western blotting of ZCCHC8 in control or ZCCHC8 knockdown i3Neurons. Arrow points the expected size of ZCCHC8. (a, b, d-g) Data are mean ± s.e.m. Points represent three biological replicates.

Source data

Extended Data Fig. 9 m6A hypomethylation of the endogenous C9 repeat-containing intron and YTHDC1 expression in the postmortem brain tissues.

a, MeRIP coupled with RT-qPCR of endogenous C9 repeat-containing intronic RNA in control and C9ORF72-ALS/FTD iPSNs. Points represent individual cell lines. n = 6 per group. *P = 0.0427. b, Representative IHC staining of YTHDC1 at temporal cortex section of control and C9ORF72-ALS/FTD samples. Scale bar = 50 μm. c, Western blotting (left) and quantification (right) of YTHDC1 using postmortem motor cortex tissue samples. n = 3 individuals per group. *Non-specific bands. *P = 0.0246. (a, c) Data are mean ± s.e.m. Points represent individual control or patient samples. P values were calculated by two-tailed t test.

Source data

Extended Data Fig. 10 m6A restoration reduces glutamate-induced excitotoxicity in C9ORF72-ALS/FTD iPSNs.

a, RT-qPCR of METTL14 in C9ORF72-ALS/FTD iPSNs expressing exogenous METTL14 or GFP control. Data are mean ± s.e.m. Points represent individual cell lines. n = 6 in each group. ***P = 0.0006 by paired two-tailed t test. b, Western blotting of FTO in C9ORF72-ALS/FTD iPSNs upon knockdown of FTO by shRNA. *non-specific band. c, d, Representative images of Hoechst and propidium iodide (PI) staining of iPSNs in the glutamate-induced excitotoxicity assay. C9ORF72-ALS/FTD iPSNs were infected with lentivirus overexpressing METTL14 or GFP as negative control (c), or expressing FTO shRNA or non-targeting control shRNA (d).

Source data

Supplementary information

Reporting Summary

41593_2023_1374_MOESM2_ESM.xlsx

Supplementary Table 1: Demographic information for lymphoblast lines. Supplementary Table 2: Demographic information for iPSC lines. Supplementary Table 3: Demographic information for frozen postmortem tissues. Supplementary Table 4: Demographic information for formalin-fixed, paraffin embedded (FFPE) tissue slides of temporal cortex. Supplementary Table 5: Sequences of qPCR primers and RT primer for antisense C9. Supplementary Table 6: Demographic information for Answer ALS and NeuroLINCS samples. Supplementary Table 7: High-throughput sequencing sample information.

Source data

Source Data Fig. 1

Statistical source data.

Source Data Fig. 2

Statistical source data.

Source Data Fig. 3

Statistical source data.

Source Data Fig. 4

Statistical source data.

Source Data Fig. 4

Unprocessed western blots.

Source Data Fig. 5

Statistical source data.

Source Data Fig. 5

Unprocessed western blots.

Source Data Fig. 6

Statistical source data.

Source Data Fig. 7

Statistical source data.

Source Data Extended Data Fig. 1

Statistical source data.

Source Data Extended Data Fig. 1

Unprocessed western blots.

Source Data Extended Data Fig. 2

Statistical source data.

Source Data Extended Data Fig. 2 and Fig. 9

Unprocessed western blots.

Source Data Extended Data Fig. 3

Statistical source data.

Source Data Extended Data Fig. 4

Statistical source data.

Source Data Extended Data Fig. 6

Statistical source data.

Source Data Extended Data Fig. 7

Statistical source data.

Source Data Extended Data Fig. 7

Unprocessed western blots.

Source Data Extended Data Fig. 8

Statistical source data.

Source Data Extended Data Fig. 8

Unprocessed western blots.

Source Data Extended Data Fig. 9

Statistical source data.

Source Data Extended Data Fig. 10

Statistical source data.

Source Data Extended Data Fig. 10

Unprocessed western blots.

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Li, Y., Dou, X., Liu, J. et al. Globally reduced N6-methyladenosine (m6A) in C9ORF72-ALS/FTD dysregulates RNA metabolism and contributes to neurodegeneration. Nat Neurosci 26, 1328–1338 (2023). https://doi.org/10.1038/s41593-023-01374-9

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41593-023-01374-9

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