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.

  • Review Article
  • Published:

Nonsense-mediated RNA decay in the brain: emerging modulator of neural development and disease

Abstract

Steady-state RNA levels are controlled by the balance between RNA synthesis and RNA turnover. A selective RNA turnover mechanism that has received recent attention in neurons is nonsense-mediated RNA decay (NMD). NMD has been shown to influence neural development, neural stem cell differentiation decisions, axon guidance and synaptic plasticity. In humans, NMD factor gene mutations cause some forms of intellectual disability and are associated with neurodevelopmental disorders, including schizophrenia and autism spectrum disorder. Impairments in NMD are linked to neurodegenerative disorders, including amyotrophic lateral sclerosis. We discuss these findings, their clinical implications and challenges for the future.

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: Nonsense-mediated RNA decay.
Fig. 2: Nonsense-mediated RNA decay-inducing features.
Fig. 3: Alternatively spliced mRNAs are often targeted by nonsense-mediated RNA decay.
Fig. 4: Nonsense-mediated RNA decay branches, regulation and neural functions.
Fig. 5: Control of protein synthesis and mRNA levels in axons by local nonsense-mediated RNA decay during axon outgrowth.
Fig. 6: Potential roles of nonsense-mediated RNA decay in suppressing neurotoxic effects of mis-spliced mRNAs.

Similar content being viewed by others

References

  1. Lennox, A. L., Mao, H. & Silver, D. L. RNA on the brain: emerging layers of post-transcriptional regulation in cerebral cortex development. Wiley Interdiscip. Rev. Dev. Biol. 7, e290 (2017).

    Google Scholar 

  2. Chang, Y.-F., Imam, J. S. & Wilkinson, M. F. The nonsense-mediated decay RNA surveillance pathway. Annu. Rev. Biochem. 76, 51–74 (2007).

    CAS  PubMed  Google Scholar 

  3. Popp, M. W.-L. & Maquat, L. E. Organizing principles of mammalian nonsense-mediated mRNA decay. Annu. Rev. Genet. 47, 139–165 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  4. Lykke-Andersen, S. & Jensen, T. H. Nonsense-mediated mRNA decay: an intricate machinery that shapes transcriptomes. Nat. Rev. Mol. Cell Biol. 16, 665–677 (2015).

    CAS  PubMed  Google Scholar 

  5. Losson, R. & Lacroute, F. Interference of nonsense mutations with eukaryotic messenger RNA stability. Proc. Natl Acad. Sci. USA 76, 5134–5137 (1979).

    CAS  PubMed  Google Scholar 

  6. Holbrook, J. A., Neu-Yilik, G., Hentze, M. W. & Kulozik, A. E. Nonsense-mediated decay approaches the clinic. Nat. Genet. 36, 801–808 (2004).

    CAS  PubMed  Google Scholar 

  7. Inoue, K. et al. Molecular mechanism for distinct neurological phenotypes conveyed by allelic truncating mutations. Nat. Genet. 36, 361–369 (2004).

    CAS  PubMed  Google Scholar 

  8. Lelivelt, M. J. & Culbertson, M. R. Yeast Upf proteins required for RNA surveillance affect global expression of the yeast transcriptome. Mol. Cell. Biol. 19, 6710–6719 (1999).

    CAS  PubMed  PubMed Central  Google Scholar 

  9. He, F. et al. Genome-wide analysis of mRNAs regulated by the nonsense-mediated and 5′ to 3′ mRNA decay pathways in yeast. Mol. Cell 12, 1439–1452 (2003).

    CAS  PubMed  Google Scholar 

  10. Mendell, J. T., Sharifi, N. A., Meyers, J. L., Martinez-Murillo, F. & Dietz, H. C. Nonsense surveillance regulates expression of diverse classes of mammalian transcripts and mutes genomic noise. Nat. Genet. 36, 1073–1078 (2004).

    CAS  PubMed  Google Scholar 

  11. Ge, Y. & Porse, B. T. The functional consequences of intron retention: alternative splicing coupled to NMD as a regulator of gene expression. Bioessays 36, 236–243 (2014).

    CAS  Google Scholar 

  12. Tani, H. et al. Identification of hundreds of novel UPF1 target transcripts by direct determination of whole transcriptome stability. RNA Biol. 9, 1370–1379 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  13. Muir, V. S., Gasch, A. P. & Anderson, P. The substrates of nonsense-mediated mRNA decay in Caenorhabditis elegans. G3 8, 195–205 (2017).

    Google Scholar 

  14. Huang, L. & Wilkinson, M. F. Regulation of nonsense-mediated mRNA decay. Wiley Interdiscip. Rev. RNA 3, 807–828 (2012).

    CAS  PubMed  Google Scholar 

  15. Karam, R., Wengrod, J., Gardner, L. L. B. & Wilkinson, M. F. M. Regulation of nonsense-mediated mRNA decay: implications for physiology and disease. Biochim. Biophys. Acta 1829, 624–633 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  16. Lou, C.-H. et al. Nonsense-mediated RNA decay influences human embryonic stem cell fate. Stem Cell Rep. 6, 844–857 (2016).

    CAS  Google Scholar 

  17. Karam, R. et al. The unfolded protein response is shaped by the NMD pathway. EMBO Rep. 16, 599–609 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  18. Li, T. et al. Smg6/Est1 licenses embryonic stem cell differentiation via nonsense-mediated mRNA decay. EMBO J. 34, 1630–1647 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  19. Nelson, J. O., Moore, K. A., Chapin, A., Hollien, J. & Metzstein, M. M. Degradation of Gadd45 mRNA by nonsense-mediated decay is essential for viability. eLife 5, e12876 (2016).

    PubMed  PubMed Central  Google Scholar 

  20. Goetz, A. E. & Wilkinson, M. Stress and the nonsense-mediated RNA decay pathway. Cell. Mol. Life Sci. 74, 3509–3531 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  21. Nagy, E. & Maquat, L. E. A rule for termination-codon position within intron-containing genes: when nonsense affects RNA abundance. Trends Biochem. Sci. 23, 198–199 (1998).

    CAS  PubMed  Google Scholar 

  22. Boehm, V. & Gehring, N. H. Exon junction complexes: supervising the gene expression assembly line. Trends Genet. 32, 724–735 (2016).

    CAS  PubMed  Google Scholar 

  23. Woodward, L. A., Mabin, J. W., Gangras, P. & Singh, G. The exon junction complex: a lifelong guardian of mRNA fate. Wiley Interdiscip. Rev. RNA 8, e1411 (2017).

    Google Scholar 

  24. Neu-Yilik, G. et al. Dual function of UPF3B in early and late translation termination. EMBO J. 36, e201797079 (2017).

    Google Scholar 

  25. Chamieh, H., Ballut, L., Bonneau, F. & Le Hir, H. NMD factors UPF2 and UPF3 bridge UPF1 to the exon junction complex and stimulate its RNA helicase activity. Nat. Struct. Mol. Biol. 15, 85–93 (2008).

    CAS  PubMed  Google Scholar 

  26. López-Perrote, A. et al. Human nonsense-mediated mRNA decay factor UPF2 interacts directly with eRF3 and the SURF complex. Nucleic Acids Res. 44, 1909–1923 (2015).

    Google Scholar 

  27. Kashima, I. et al. Binding of a novel SMG-1-Upf1-eRF1-eRF3 complex (SURF) to the exon junction complex triggers Upf1 phosphorylation and nonsense-mediated mRNA decay. Genes Dev. 20, 355–367 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  28. Dostie, J. & Dreyfuss, G. Translation is required to remove Y14 from mRNAs in the cytoplasm. Curr. Biol. 12, 1060–1067 (2002).

    CAS  PubMed  Google Scholar 

  29. Zünd, D., Gruber, A. R., Zavolan, M. & Mühlemann, O. Translation-dependent displacement of UPF1 from coding sequences causes its enrichment in 3’ UTRs. Nat. Struct. Mol. Biol. 20, 936–943 (2013).

    PubMed  Google Scholar 

  30. Hurt, J. A., Robertson, A. D. & Burge, C. B. Global analyses of UPF1 binding and function reveal expanded scope of nonsense-mediated mRNA decay. Genome Res. 23, 1636–1650 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  31. Tarpey, P. S. et al. Mutations in UPF3B, a member of the nonsense-mediated mRNA decay complex, cause syndromic and nonsyndromic mental retardation. Nat. Genet. 39, 1127–1133 (2007). This report is the first to demonstrate that UPF3B mutations are likely to cause ID in humans. The authors also find that patients with ID with UPF3B mutations sometimes have neurodevelopmental disorders, such as schizophrenia.

    CAS  PubMed  PubMed Central  Google Scholar 

  32. Laumonnier, F. et al. Mutations of the UPF3B gene, which encodes a protein widely expressed in neurons, are associated with nonspecific mental retardation with or without autism. Mol. Psychiatry 15, 767–776 (2010).

    CAS  PubMed  Google Scholar 

  33. Addington, A. M. et al. A novel frameshift mutation in UPF3B identified in brothers affected with childhood onset schizophrenia and autism spectrum disorders. Mol. Psychiatry 16, 238–239 (2011).

    CAS  PubMed  Google Scholar 

  34. Lynch, S. A. et al. Broadening the phenotype associated with mutations in UPF3B: two further cases with renal dysplasia and variable developmental delay. Eur. J. Med. Genet. 55, 476–479 (2012).

    PubMed  Google Scholar 

  35. Xu, X. et al. Exome sequencing identifies UPF3B as the causative gene for a Chinese non-syndrome mental retardation pedigree. Clin. Genet. 83, 560–564 (2012).

    PubMed  Google Scholar 

  36. Huang, L. et al. A Upf3b-mutant mouse model with behavioral and neurogenesis defects. Mol. Psychiatry https://doi.org/10.1038/mp.2017.173 (2017). This study shows that mice harbouring loss-of-function mutations in Upf3b have specific behavioural defects and reduced dendritic spine maturation in specific brain regions. Cultured neural progenitors from these NMD-deficient mice exhibit perturbed neural differentiation

    PubMed  PubMed Central  Google Scholar 

  37. Chan, W.-K. et al. An alternative branch of the nonsense-mediated decay pathway. EMBO J. 26, 1820–1830 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  38. Huang, L. et al. RNA homeostasis governed by cell type-specific and branched feedback loops acting on NMD. Mol. Cell 43, 950–961 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  39. Medghalchi, S. M. et al. Rent1, a trans-effector of nonsense-mediated mRNA decay, is essential for mammalian embryonic viability. Hum. Mol. Genet. 10, 99–105 (2001).

    CAS  PubMed  Google Scholar 

  40. Weischenfeldt, J. et al. NMD is essential for hematopoietic stem and progenitor cells and for eliminating by-products of programmed DNA rearrangements. Genes Dev. 22, 1381–1396 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  41. McIlwain, D. R. et al. Smg1 is required for embryogenesis and regulates diverse genes via alternative splicing coupled to nonsense-mediated mRNA decay. Proc. Natl Acad. Sci. USA 107, 12186–12191 (2010).

    PubMed  Google Scholar 

  42. Shum, E. Y. et al. The antagonistic gene paralogs Upf3a and Upf3b govern nonsense-mediated RNA decay. Cell 165, 382–395 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  43. Nguyen, L. S. et al. Contribution of copy number variants involving nonsense-mediated mRNA decay pathway genes to neuro-developmental disorders. Hum. Mol. Genet. 22, 1816–1825 (2013).

    CAS  PubMed  Google Scholar 

  44. Gulsuner, S. et al. Spatial and temporal mapping of de novo mutations in schizophrenia to a fetal prefrontal cortical network. Cell 154, 518–529 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  45. Brunetti-Pierri, N. et al. Recurrent reciprocal 1q21.1 deletions and duplications associated with microcephaly or macrocephaly and developmental and behavioral abnormalities. Nat. Genet. 40, 1466–1471 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  46. Mefford, H. C. et al. Recurrent rearrangements of chromosome 1q21.1 and variable pediatric phenotypes. N. Engl. J. Med. 359, 1685–1699 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  47. Rosenfeld, J. A. et al. Proximal microdeletions and microduplications of 1q21.1 contribute to variable abnormal phenotypes. Eur. J. Hum. Genet. 20, 754–761 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  48. Mao, H. et al. Rbm8a haploinsufficiency disrupts embryonic cortical development resulting in microcephaly. J. Neurosci. 35, 7003–7018 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  49. Mao, H., McMahon, J. J., Tsai, Y.-H., Wang, Z. & Silver, D. L. Haploinsufficiency for core exon junction complex components disrupts embryonic neurogenesis and causes p53-mediated microcephaly. PLOS Genet. 12, e1006282 (2016).

    PubMed  PubMed Central  Google Scholar 

  50. Alachkar, A. et al. An EJC factor RBM8a regulates anxiety behaviors. Curr. Mol. Med. 13, 887–899 (2013).

    CAS  PubMed  Google Scholar 

  51. Singh, G. et al. The cellular EJC interactome reveals higher-order mRNP structure and an EJC-SR protein nexus. Cell 151, 750–764 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  52. Saulière, J. et al. CLIP-seq of eIF4AIII reveals transcriptome-wide mapping of the human exon junction complex. Nat. Struct. Mol. Biol. 19, 1124–1131 (2012).

    PubMed  Google Scholar 

  53. Bruno, I. G. et al. Identification of a microRNA that activates gene expression by repressing nonsense-mediated RNA decay. Mol. Cell 42, 500–510 (2011). This study identifies the first miRNA that regulates NMD. This miRNA — miR-128 — is induced during neural differentiation, where it functions to downregulate NMD.

    CAS  PubMed  PubMed Central  Google Scholar 

  54. Jolly, L. A., Homan, C. C., Jacob, R., Barry, S. & Gecz, J. The UPF3B gene, implicated in intellectual disability, autism, ADHD and childhood onset schizophrenia regulates neural progenitor cell behaviour and neuronal outgrowth. Hum. Mol. Genet. 22, 4673–4687 (2013). Using a knockdown approach, this study shows that Upf3b influences neural progenitor proliferation versus differentiation decisions, as well as neurite outgrowth. This study also demonstrates that Upf3a and Upf3b exhibit interesting patterns of expression during brain development.

    CAS  PubMed  Google Scholar 

  55. Lou, C. H. et al. Posttranscriptional control of the stem cell and neurogenic programs by the nonsense-mediated RNA decay pathway. Cell Rep. 6, 748–764 (2014). This study provides evidence for an NMD-based molecular circuit that controls neural differentiation. Neurally expressed miRNAs downregulate NMD, which, in turn, upregulate pro-neural and anti-proliferation proteins encoded by NMD target mRNAs.

    CAS  PubMed  PubMed Central  Google Scholar 

  56. Alrahbeni, T. et al. Full UPF3B function is critical for neuronal differentiation of neural stem cells. Mol. Brain 8, 33 (2015).

    PubMed  PubMed Central  Google Scholar 

  57. Cho, H. et al. Staufen1-mediated mRNA decay functions in adipogenesis. Mol. Cell 46, 495–506 (2012).

    CAS  PubMed  Google Scholar 

  58. Gong, C., Kim, Y. K., Woeller, C. F., Tang, Y. & Maquat, L. E. SMD and NMD are competitive pathways that contribute to myogenesis: effects on PAX3 and myogenin mRNAs. Genes Dev. 23, 54–66 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  59. Wang, G., Jiang, B., Jia, C., Chai, B. & Liang, A. MicroRNA 125 represses nonsense-mediated mRNA decay by regulating SMG1 expression. Biochem. Biophys. Res. Commun. 435, 16–20 (2013).

    CAS  PubMed  Google Scholar 

  60. Akerblom, M. et al. microRNA-125 distinguishes developmentally generated and adult-born olfactory bulb interneurons. Development 141, 1580–1588 (2014).

    CAS  PubMed  Google Scholar 

  61. Lattanzi, A. et al. Dynamic activity of miR-125b and miR-93 during murine neural stem cell differentiation in vitro and in the subventricular zone neurogenic niche. PLOS ONE 8, e67411 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  62. Yan, Q. et al. Systematic discovery of regulated and conserved alternative exons in the mammalian brain reveals NMD modulating chromatin regulators. Proc. Natl Acad. Sci. USA 112, 3445–3450 (2015). This study identifies hundreds of alternatively spliced NMD-inducing exons expressed in the mouse brain cortex. These ‘NMD exons’ are enriched in genes encoding RNA-binding proteins and chromatin regulators, suggesting the existence of a gene regulatory feedback loop operating in the cortex.

    CAS  PubMed  Google Scholar 

  63. Krey, J. F. et al. Timothy syndrome is associated with activity-dependent dendritic retraction in rodent and human neurons. Nat. Neurosci. 16, 201–209 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  64. Nguyen, L. S. et al. Transcriptome profiling of UPF3B/NMD-deficient lymphoblastoid cells from patients with various forms of intellectual disability. Mol. Psychiatry 17, 1103–1115 (2012).

    CAS  PubMed  Google Scholar 

  65. Wittkopp, N. et al. Nonsense-mediated mRNA decay effectors are essential for zebrafish embryonic development and survival. Mol. Cell. Biol. 29, 3517–3528 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  66. Silver, D. L. et al. The exon junction complex component Magoh controls brain size by regulating neural stem cell division. Nat. Neurosci. 13, 551–558 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  67. Pilaz, L. et al. Prolonged mitosis of neural progenitors alters cell fate in the developing brain. Neuron 89, 83–99 (2016). Using mice haploid for the EJC factor gene Magoh, this study finds that Magoh is critical for developing radial glia, including their survival, the timing at which they initiate mitosis and the number of cell divisions that they undergo.

    CAS  PubMed  PubMed Central  Google Scholar 

  68. Zou, D. et al. A critical role of RBM8a in proliferation and differentiation of embryonic neural progenitors. Neural Dev. 10, 18 (2015).

    PubMed  PubMed Central  Google Scholar 

  69. Chen, Z., Gore, B. B., Long, H., Ma, L. & Tessier-Lavigne, M. Alternative splicing of the Robo3 axon guidance receptor governs the midline switch from attraction to repulsion. Neuron 58, 325–332 (2008).

    CAS  PubMed  Google Scholar 

  70. Jaworski, A., Long, H. & Tessier-Lavigne, M. Collaborative and specialized functions of Robo1 and Robo2 in spinal commissural axon guidance. J. Neurosci. 30, 9445–9453 (2010).

    CAS  PubMed  Google Scholar 

  71. Long, H. et al. Conserved roles for Slit and Robo proteins in midline commissural axon guidance. Neuron 42, 213–223 (2004).

    CAS  PubMed  Google Scholar 

  72. Sabatier, C. et al. The divergent robo family protein Rig-1/Robo3 is a negative regulator of slit responsiveness required for midline crossing by commissural axons. Cell 117, 157–169 (2004).

    CAS  PubMed  Google Scholar 

  73. Colak, D., Ji, S.-J., Porse, B. T. & Jaffrey, S. R. Regulation of axon guidance by compartmentalized nonsense-mediated mRNA decay. Cell 153, 1252–1265 (2013). This study reveals the existence of ‘local’ NMD occurring within axon terminals that controls the mRNA composition of pathfinding axons. NMD was found to target Robo3.2 mRNA, thereby controlling the expression this important axon guidance protein.

    CAS  PubMed  PubMed Central  Google Scholar 

  74. Maquat, L. E., Tarn, W. Y. & Isken, O. The pioneer round of translation: features and functions. Cell 142, 368–374 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  75. Ivanov, P. V., Gehring, N. H., Kunz, J. B., Hentze, M. W. & Kulozik, A. E. Interactions between UPF1, eRFs, PABP and the exon junction complex suggest an integrated model for mammalian NMD pathways. EMBO J. 27, 736–747 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  76. Mills, E. W., Wangen, J., Green, R. & Ingolia, N. T. Dynamic regulation of a ribosome rescue pathway in erythroid cells and platelets. Cell Rep. 17, 1–10 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  77. Rufener, S. C. & Mühlemann, O. EIF4E-bound mRNPs are substrates for nonsense-mediated mRNA decay in mammalian cells. Nat. Struct. Mol. Biol. 20, 710–717 (2013).

    CAS  PubMed  Google Scholar 

  78. Durand, S. & Lykke-Andersen, J. Nonsense-mediated mRNA decay occurs during eIF4F-dependent translation in human cells. Nat. Struct. Mol. Biol. 20, 702–709 (2013).

    CAS  PubMed  Google Scholar 

  79. Kiebler, M. A. & Bassell, G. J. Neuronal RNA granules: movers and makers. Neuron 51, 685–690 (2006).

    CAS  PubMed  Google Scholar 

  80. Carter, M. S. et al. A regulatory mechanism that detects premature nonsense codons in T cell receptor transcripts in vivo is reversed by protein synthesis inhibitors in vitro. J. Biol. Chem. 270, 28995–29003 (1995).

    CAS  PubMed  Google Scholar 

  81. Giorgi, C. et al. The EJC factor eIF4AIII modulates synaptic strength and neuronal protein expression. Cell 130, 179–191 (2007). This study links synaptic activity to NMD-dependent degradation of the Arc mRNA, a natural NMD target that encodes a key synaptic regulator. This study shows that NMD factor components are present at synapses, providing the first hints that NMD can function at synaptic sites.

    CAS  PubMed  Google Scholar 

  82. Lyford, G. L. et al. Arc, a growth factor and activity-regulated gene, encodes a novel cytoskeleton-associated protein that is enriched in neuronal dendrites. Neuron 14, 433–445 (1995).

    CAS  PubMed  Google Scholar 

  83. Link, W. W. et al. Somatodendritic expression of an immediate early gene is regulated by synaptic activity. Proc. Natl Acad. Sci. USA 92, 5734–5738 (1995).

    CAS  PubMed  Google Scholar 

  84. Farris, S., Lewandowski, G., Cox, C. D. & Steward, O. Selective localization of Arc mRNA in dendrites involves activity- and translation-dependent mRNA degradation. J. Neurosci. 34, 4481–4493 (2014). This study reveals evidence that NMD controls the levels of the Arc transcript at synapses, which encodes a key synaptic regulator.

    PubMed  PubMed Central  Google Scholar 

  85. Paolantoni, C. et al. Arc 3’ UTR splicing leads to dual and antagonistic effects in fine-tuning Arc expression upon BDNF signaling. Front. Mol. Neurosci. 11, 145 (2018).

    PubMed  PubMed Central  Google Scholar 

  86. Guzowski, J. F. et al. Mapping behaviorally relevant neural circuits with immediate-early gene expression. Curr. Opin. Neurobiol. 15, 599–606 (2005).

    CAS  PubMed  Google Scholar 

  87. Alonso, C. R. A complex ‘mRNA degradation code’ controls gene expression during animal development. Trends Genet. 28, 78–88 (2012).

    CAS  PubMed  Google Scholar 

  88. Jaagura, M. et al. Rat NEURL1 3’UTR is alternatively spliced and targets mRNA to dendrites. Neurosci. Lett. 635, 71–76 (2016).

    CAS  PubMed  Google Scholar 

  89. Li, Y. et al. An NXF1 mRNA with a retained intron is expressed in hippocampal and neocortical neurons and is translated into a protein that functions as an Nxf1 cofactor. Mol. Biol. Cell 27, 3903–3912 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  90. Long, A. A. et al. The nonsense-mediated decay pathway maintains synapse architecture and synaptic vesicle cycle efficacy. J. Cell Sci. 123, 3303–3315 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  91. Zheng, S. et al. PSD-95 is post-transcriptionally repressed during early neural development by PTBP1 and PTBP2. Nat. Neurosci. 15, 381–8, S1 (2012). This study provides evidence that NMD is critical for the regulation of a factor that mediates synaptic maturation and plasticity.

    CAS  PubMed  PubMed Central  Google Scholar 

  92. Zheng, S. Alternative splicing and nonsense-mediated mRNA decay enforce neural specific gene expression. Int. J. Dev. Neurosci. 55, 102–108 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  93. Ju, S. et al. A yeast model of FUS/TLS-dependent cytotoxicity. PLOS Biol. 9, e1001052 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  94. Sun, Z. et al. Molecular determinants and genetic modifiers of aggregation and toxicity for the ALS disease protein FUS/TLS. PLOS Biol. 9, e1000614 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  95. Jackson, K. L. et al. Preservation of forelimb function by UPF1 gene therapy in a rat model of TDP-43-induced motor paralysis. Gene Ther. 22, 20–28 (2015). Using a rat model of ALS, the investigators of this study show that overexpression of the NMD protein UPF1 can reduce symptoms of ALS, including motor paralysis.

    CAS  PubMed  PubMed Central  Google Scholar 

  96. Barmada, S. J. et al. Amelioration of toxicity in neuronal models of amyotrophic lateral sclerosis by hUPF1. Proc. Natl Acad. Sci. USA 112, 7821–7826 (2015). This study shows that overexpression of the NMD factor UPF1 reduces toxicity in a neuronal model of ALS.

    CAS  PubMed  Google Scholar 

  97. Gregersen, L. H. et al. MOV10 Is a 5′ to 3′ RNA helicase contributing to UPF1 mRNA target degradation by translocation along 3′ UTRs. Mol. Cell 54, 573–585 (2014).

    CAS  PubMed  Google Scholar 

  98. Conlon, E. G. & Manley, J. L. RNA-binding proteins in neurodegeneration: mechanisms in aggregate. Genes Dev. 31, 1509–1528 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  99. Qiu, H. et al. ALS-associated mutation FUS-R521C causes DNA damage and RNA splicing defects. J. Clin. Invest. 124, 981–999 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  100. Miller, J. W. Recruitment of human muscleblind proteins to (CUG)n expansions associated with myotonic dystrophy. EMBO J. 19, 4439–4448 (2000).

    CAS  PubMed  PubMed Central  Google Scholar 

  101. Lotti, F. et al. An SMN-dependent U12 splicing event essential for motor circuit function. Cell 151, 440–454 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  102. Nickless, A. et al. Intracellular calcium regulates nonsense-mediated mRNA decay. Nat. Med. 20, 961–966 (2014).

    CAS  PubMed  Google Scholar 

  103. Nickless, A., Bailis, J. M. & You, Z. Control of gene expression through the nonsense-mediated RNA decay pathway. Cell Biosci. 7, 26 (2017).

    PubMed  PubMed Central  Google Scholar 

  104. Wilkinson, M. F. The cycle of nonsense. Mol. Cell 12, 1059–1061 (2003).

    CAS  PubMed  Google Scholar 

  105. Melero, R. et al. The RNA helicase DHX34 functions as a scaffold for SMG1-mediated UPF1 phosphorylation. Nat. Commun. 7, 10585 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  106. Phelps, S. F. et al. Expression of full-length and truncated dystrophin mini-genes in transgenic mdx mice. Hum. Mol. Genet. 4, 1251–1258 (1995).

    CAS  PubMed  Google Scholar 

  107. Yuasa, K. et al. Effective restoration of dystrophin-associated proteins in vivo by adenovirus-mediated transfer of truncated dystrophin cDNAs. FEBS Lett. 425, 329–336 (1998).

    CAS  PubMed  Google Scholar 

  108. Durand, S. et al. Inhibition of nonsense-mediated mRNA decay (NMD) by a new chemical molecule reveals the dynamic of NMD factors in P-bodies. J. Cell Biol. 178, 1145–1160 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  109. Martin, L. et al. Identification and characterization of small molecules that inhibit nonsense-mediated RNA decay and suppress nonsense p53 mutations. Cancer Res. 74, 3104–3113 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  110. Ge, Z., Quek, B. L., Beemon, K. L. & Hogg, J. R. Polypyrimidine tract binding protein 1 protects mRNAs from recognition by the nonsense-mediated mRNA decay pathway. eLife 5, 1–25 (2016).

    Google Scholar 

  111. Coe, B. P. et al. Refining analyses of copy number variation identifies specific genes associated with developmental delay. Nat. Genet. 46, 1063–1071 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  112. Favaro, F. P. et al. A noncoding expansion in EIF4A3 causes Richieri-Costa-Pereira syndrome, a craniofacial disorder associated with limb defects. Am. J. Hum. Genet. 94, 120–128 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

This work was supported by US National Institute of Health grants NS056306 (S.J.) and R01 GM111838 (M.F.W.).

Reviewer information

Nature Reviews Neuroscience thanks D. Silver and the other anonymous reviewers for their contribution to the peer review of this work.

Author information

Authors and Affiliations

Authors

Contributions

S.J. and M.F.W. researched data for the article and made substantial contributions to discussion of content and the writing, review and editing of the manuscript before submission.

Corresponding authors

Correspondence to Samie R. Jaffrey or Miles F. Wilkinson.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Publisher’s note

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

Glossary

RNA turnover

Degradation of RNAs; typically achieved through the action of specific nucleases.

RNA surveillance

The recognition and subsequent elimination of abnormal RNAs.

Dominant-negative

Mutant proteins that antagonize the function of the wild-type protein.

Transcripts

RNAs, including those coding for protein (mRNAs).

Genome-wide studies

Analyses of the majority of the genes in a genome (for example, RNA sequencing analysis determines the level of RNAs transcribed from all significantly expressed genes in the genome).

In-frame

Codons that are in the same frame as the initiator codon.

Untranslated regions

(UTRs). The regions of an mRNA upstream and downstream of the coding region (that is, the 5′ UTR is upstream of the initiator codon and the 3′ UTR is downstream of the stop codon).

Haploinsufficiency

Loss of one copy of a gene from a diploid organism.

Commissural axons

Neurites (projections from the cell body) that cross the midline of the CNS to the other side of the nervous system.

Frameshift

Insertions and deletions downstream of the initiator codon that are not a multiple of three and thus shift the reading frame (this leads to altered amino acids in the encoded protein downstream of the frameshift).

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Jaffrey, S.R., Wilkinson, M.F. Nonsense-mediated RNA decay in the brain: emerging modulator of neural development and disease. Nat Rev Neurosci 19, 715–728 (2018). https://doi.org/10.1038/s41583-018-0079-z

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41583-018-0079-z

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