Skip to main content

Lost in Translation: Evidence for Protein Synthesis Deficits in ALS/FTD and Related Neurodegenerative Diseases

  • Chapter
  • First Online:
RNA Metabolism in Neurodegenerative Diseases

Part of the book series: Advances in Neurobiology ((NEUROBIOL,volume 20))

Abstract

Cells utilize a complex network of proteins to regulate translation, involving post-transcriptional processing of RNA and assembly of the ribosomal unit. Although the complexity provides robust regulation of proteostasis, it also offers several opportunities for translational dysregulation, as has been observed in many neurodegenerative disorders. Defective mRNA localization, mRNA sequatration, inhibited ribogenesis, mutant tRNA synthetases, and translation of hexanucleotide expansions have all been associated with neurodegenerative disease. Here, we review dysregulation of translation in the context of age-related neurodegeneration and discuss novel methods to interrogate translation. This review primarily focuses on amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD), a spectrum disorder heavily associated with RNA metabolism, while also analyzing translational inhibition in the context of related neurodegenerative disorders such as Alzheimer’s disease and Huntington’s disease and the translation-related pathomechanisms common in neurodegenerative disease.

This is a preview of subscription content, log in via an institution to check access.

Access this chapter

Chapter
USD 29.95
Price excludes VAT (USA)
  • Available as PDF
  • Read on any device
  • Instant download
  • Own it forever
eBook
USD 84.99
Price excludes VAT (USA)
  • Available as EPUB and PDF
  • Read on any device
  • Instant download
  • Own it forever
Softcover Book
USD 109.99
Price excludes VAT (USA)
  • Compact, lightweight edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info
Hardcover Book
USD 109.99
Price excludes VAT (USA)
  • Durable hardcover edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info

Tax calculation will be finalised at checkout

Purchases are for personal use only

Institutional subscriptions

References

  1. Aitken CE, Lorsch JR. A mechanistic overview of translation initiation in eukaryotes. Nat Struct Mol Biol. 2012;19(6):568–76.

    Article  PubMed  CAS  Google Scholar 

  2. Komar AA, Hatzoglou M. Cellular IRES-mediated translation: the war of ITAFs in pathophysiological states. Cell Cycle. 2011;10(2):229–40.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  3. Veo BL, Krushel LA. Translation initiation of the human tau mRNA through an internal ribosomal entry site. J Alzheimers Dis. 2009;16(2):271–5.

    Article  PubMed  CAS  Google Scholar 

  4. Cleary JD, Ranum LP. New developments in RAN translation: insights from multiple diseases. Curr Opin Genet Dev. 2017;44:125–34.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  5. Gao FB, Richter JD, Cleveland DW. Rethinking unconventional translation in neurodegeneration. Cell. 2017;171(5):994–1000.

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  6. Zu T, Gibbens B, Doty NS, Gomes-Pereira M, Huguet A, Stone MD, et al. Non-ATG-initiated translation directed by microsatellite expansions. Proc Natl Acad Sci U S A. 2011;108(1):260–5.

    Article  PubMed  Google Scholar 

  7. Freibaum BD, Taylor JP. The role of dipeptide repeats in C9ORF72-related ALS-FTD. Front Mol Neurosci. 2017;10:35.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  8. Boczonadi V, Horvath R. Mitochondria: impaired mitochondrial translation in human disease. Int J Biochem Cell Biol. 2014;48:77–84.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  9. Scheper GC, van der Knaap MS, Proud CG. Translation matters: protein synthesis defects in inherited disease. Nat Rev Genet. 2007;8(9):711–23.

    Article  PubMed  CAS  Google Scholar 

  10. van der Knaap MS, Leegwater PA, Konst AA, Visser A, Naidu S, Oudejans CB, et al. Mutations in each of the five subunits of translation initiation factor eIF2B can cause leukoencephalopathy with vanishing white matter. Ann Neurol. 2002;51(2):264–70.

    Article  PubMed  CAS  Google Scholar 

  11. Antonellis A, Ellsworth RE, Sambuughin N, Puls I, Abel A, Lee-Lin SQ, et al. Glycyl tRNA synthetase mutations in Charcot-Marie-Tooth disease type 2D and distal spinal muscular atrophy type V. Am J Hum Genet. 2003;72(5):1293–9.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  12. Chen E, Sharma MR, Shi X, Agrawal RK, Joseph S. Fragile x mental retardation protein regulates translation by binding directly to the ribosome. Mol Cell. 2014;54(3):407–17.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  13. Kubik-Zahorodna A, Schuster B, Kanchev I, Sedlacek R. Neurological deficits of an Rps19(Arg67del) model of Diamond-Blackfan anaemia. Folia Biol (Praha). 2016;62(4):139–47.

    CAS  Google Scholar 

  14. Steenweg ME, Ghezzi D, Haack T, Abbink TE, Martinelli D, van Berkel CG, et al. Leukoencephalopathy with thalamus and brainstem involvement and high lactate ‘LTBL’ caused by EARS2 mutations. Brain. 2012;135(Pt 5):1387–94.

    Article  PubMed  Google Scholar 

  15. Coyne AN, Lorenzini I, Chou CC, Torvund M, Rogers RS, Starr A, et al. Post-transcriptional inhibition of Hsc70-4/HSPA8 expression leads to synaptic vesicle cycling defects in multiple models of ALS. Cell Rep. 2017;21(1):110–25.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  16. Hebert SS, Horre K, Nicolai L, Papadopoulou AS, Mandemakers W, Silahtaroglu AN, et al. Loss of microRNA cluster miR-29a/b-1 in sporadic Alzheimer's disease correlates with increased BACE1/beta-secretase expression. Proc Natl Acad Sci U S A. 2008;105(17):6415–20.

    Article  PubMed  PubMed Central  Google Scholar 

  17. Jovicic A, Mertens J, Boeynaems S, Bogaert E, Chai N, Yamada SB, et al. Modifiers of C9orf72 dipeptide repeat toxicity connect nucleocytoplasmic transport defects to FTD/ALS. Nat Neurosci. 2015;18(9):1226–9.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  18. Cleveland DW, Rothstein JD. From Charcot to Lou Gehrig: deciphering selective motor neuron death in ALS. Nat Rev Neurosci. 2001;2(11):806–19.

    Article  PubMed  CAS  Google Scholar 

  19. Schymick JC, Traynor BJ. Expanding the genetics of amyotrophic lateral sclerosis and frontotemporal dementia. Alzheimers Res Ther. 2012;4(4):30.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  20. Ling SC, Polymenidou M, Cleveland DW. Converging mechanisms in ALS and FTD: disrupted RNA and protein homeostasis. Neuron. 2013;79(3):416–38.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  21. Bruijn LI, Houseweart MK, Kato S, Anderson KL, Anderson SD, Ohama E, et al. Aggregation and motor neuron toxicity of an ALS-linked SOD1 mutant independent from wild-type SOD1. Science. 1998;281(5384):1851–4.

    Article  PubMed  CAS  Google Scholar 

  22. Tafuri F, Ronchi D, Magri F, Comi GP, Corti S. SOD1 misplacing and mitochondrial dysfunction in amyotrophic lateral sclerosis pathogenesis. Front Cell Neurosci. 2015;9:336.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  23. Tan W, Naniche N, Bogush A, Pedrini S, Trotti D, Pasinelli P. Small peptides against the mutant SOD1/Bcl-2 toxic mitochondrial complex restore mitochondrial function and cell viability in mutant SOD1-mediated ALS. J Neurosci. 2013;33(28):11588–98.

    Article  PubMed  CAS  Google Scholar 

  24. Hetz C, Mollereau B. Disturbance of endoplasmic reticulum proteostasis in neurodegenerative diseases. Nat Rev Neurosci. 2014;15(4):233–49.

    Article  PubMed  CAS  Google Scholar 

  25. Gal J, Kuang L, Barnett KR, Zhu BZ, Shissler SC, Korotkov KV, et al. ALS mutant SOD1 interacts with G3BP1 and affects stress granule dynamics. Acta Neuropathol. 2016;132(4):563–76.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  26. Kedersha N, Anderson P. Regulation of translation by stress granules and processing bodies. Prog Mol Biol Transl Sci. 2009;90:155–85.

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  27. Sun S, Sun Y, Ling SC, Ferraiuolo L, McAlonis-Downes M, Zou Y, et al. Translational profiling identifies a cascade of damage initiated in motor neurons and spreading to glia in mutant SOD1-mediated ALS. Proc Natl Acad Sci U S A. 2015;112(50):E6993–7002.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  28. Harding HP, Calfon M, Urano F, Novoa I, Ron D. Transcriptional and translational control in the Mammalian unfolded protein response. Annu Rev Cell Dev Biol. 2002;18:575–99.

    Article  PubMed  CAS  Google Scholar 

  29. Buratti E, Dork T, Zuccato E, Pagani F, Romano M, Baralle FE. Nuclear factor TDP-43 and SR proteins promote in vitro and in vivo CFTR exon 9 skipping. EMBO J. 2001;20(7):1774–84.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  30. Neumann M, Sampathu DM, Kwong LK, Truax AC, Micsenyi MC, Chou TT, et al. Ubiquitinated TDP-43 in frontotemporal lobar degeneration and amyotrophic lateral sclerosis. Science. 2006;314(5796):130–3.

    Article  CAS  PubMed  Google Scholar 

  31. Ishiguro A, Kimura N, Watanabe Y, Watanabe S, Ishihama A. TDP-43 binds and transports G-quadruplex-containing mRNAs into neurites for local translation. Genes Cells. 2016;21(5):466–81.

    Article  PubMed  CAS  Google Scholar 

  32. Coyne AN, Siddegowda BB, Estes PS, Johannesmeyer J, Kovalik T, Daniel SG, et al. Futsch/MAP1B mRNA is a translational target of TDP-43 and is neuroprotective in a Drosophila model of amyotrophic lateral sclerosis. J Neurosci. 2014;34(48):15962–74.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  33. Alami NH, Smith RB, Carrasco MA, Williams LA, Winborn CS, Han SS, et al. Axonal transport of TDP-43 mRNA granules is impaired by ALS-causing mutations. Neuron. 2014;81(3):536–43.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  34. Voigt A, Herholz D, Fiesel FC, Kaur K, Muller D, Karsten P, et al. TDP-43-mediated neuron loss in vivo requires RNA-binding activity. PLoS One. 2010;5(8):e12247.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  35. Estes PS, Boehringer A, Zwick R, Tang JE, Grigsby B, Zarnescu DC. Wild-type and A315T mutant TDP-43 exert differential neurotoxicity in a Drosophila model of ALS. Hum Mol Genet. 2011;20(12):2308–21.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  36. Estes PS, Daniel SG, McCallum AP, Boehringer AV, Sukhina AS, Zwick RA, et al. Motor neurons and glia exhibit specific individualized responses to TDP-43 expression in a Drosophila model of amyotrophic lateral sclerosis. Dis Model Mech. 2013;6(3):721–33.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  37. Majumder P, Chen YT, Bose JK, Wu CC, Cheng WC, Cheng SJ, et al. TDP-43 regulates the mammalian spinogenesis through translational repression of Rac1. Acta Neuropathol. 2012;124(2):231–45.

    Article  PubMed  CAS  Google Scholar 

  38. Liu T, Daniels CK, Cao S. Comprehensive review on the HSC70 functions, interactions with related molecules and involvement in clinical diseases and therapeutic potential. Pharmacol Ther. 2012;136(3):354–74.

    Article  PubMed  CAS  Google Scholar 

  39. Sephton CF, Cenik C, Kucukural A, Dammer EB, Cenik B, Han Y, et al. Identification of neuronal RNA targets of TDP-43-containing ribonucleoprotein complexes. J Biol Chem. 2011;286(2):1204–15.

    Article  PubMed  CAS  Google Scholar 

  40. Freibaum BD, Chitta RK, High AA, Taylor JP. Global analysis of TDP-43 interacting proteins reveals strong association with RNA splicing and translation machinery. J Proteome Res. 2010;9(2):1104–20.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  41. Majumder P, Chu JF, Chatterjee B, Swamy KB, Shen CJ. Co-regulation of mRNA translation by TDP-43 and Fragile X Syndrome protein FMRP. Acta Neuropathol. 2016;132(5):721–38.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  42. Coyne AN, Yamada SB, Siddegowda BB, Estes PS, Zaepfel BL, Johannesmeyer JS, et al. Fragile X protein mitigates TDP-43 toxicity by remodeling RNA granules and restoring translation. Hum Mol Genet. 2015;24(24):6886–98.

    PubMed  PubMed Central  CAS  Google Scholar 

  43. Wang JW, Brent JR, Tomlinson A, Shneider NA, McCabe BD. The ALS-associated proteins FUS and TDP-43 function together to affect Drosophila locomotion and life span. J Clin Invest. 2011;121(10):4118–26.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  44. Fiesel FC, Weber SS, Supper J, Zell A, Kahle PJ. TDP-43 regulates global translational yield by splicing of exon junction complex component SKAR. Nucleic Acids Res. 2012;40(6):2668–82.

    Article  PubMed  CAS  Google Scholar 

  45. Ma XM, Yoon SO, Richardson CJ, Julich K, Blenis J. SKAR links pre-mRNA splicing to mTOR/S6K1-mediated enhanced translation efficiency of spliced mRNAs. Cell. 2008;133(2):303–13.

    Article  PubMed  CAS  Google Scholar 

  46. Kim HJ, Raphael AR, LaDow ES, McGurk L, Weber RA, Trojanowski JQ, et al. Therapeutic modulation of eIF2alpha phosphorylation rescues TDP-43 toxicity in amyotrophic lateral sclerosis disease models. Nat Genet. 2014;46(2):152–60.

    Article  PubMed  CAS  Google Scholar 

  47. Vance C, Rogelj B, Hortobagyi T, De Vos KJ, Nishimura AL, Sreedharan J, et al. Mutations in FUS, an RNA processing protein, cause familial amyotrophic lateral sclerosis type 6. Science. 2009;323(5918):1208–11.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  48. Yamaguchi A, Takanashi K. FUS interacts with nuclear matrix-associated protein SAFB1 as well as Matrin3 to regulate splicing and ligand-mediated transcription. Sci Rep. 2016;6:35195.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  49. DeJesus-Hernandez M, Mackenzie IR, Boeve BF, Boxer AL, Baker M, Rutherford NJ, et al. Expanded GGGGCC hexanucleotide repeat in noncoding region of C9ORF72 causes chromosome 9p-linked FTD and ALS. Neuron. 2011;72(2):245–56.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  50. Renton AE, Majounie E, Waite A, Simon-Sanchez J, Rollinson S, Gibbs JR, et al. A hexanucleotide repeat expansion in C9ORF72 is the cause of chromosome 9p21-linked ALS-FTD. Neuron. 2011;72(2):257–68.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  51. van Blitterswijk M, van Es MA, Hennekam EA, Dooijes D, van Rheenen W, Medic J, et al. Evidence for an oligogenic basis of amyotrophic lateral sclerosis. Hum Mol Genet. 2012;21(17):3776–84.

    Article  PubMed  CAS  Google Scholar 

  52. Levine TP, Daniels RD, Gatta AT, Wong LH, Hayes MJ. The product of C9orf72, a gene strongly implicated in neurodegeneration, is structurally related to DENN Rab-GEFs. Bioinformatics. 2013;29(4):499–503.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  53. Gitler AD, Tsuiji H. There has been an awakening: emerging mechanisms of C9orf72 mutations in FTD/ALS. Brain Res. 2016;1647:19–29.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  54. Freibaum BD, Lu Y, Lopez-Gonzalez R, Kim NC, Almeida S, Lee KH, et al. GGGGCC repeat expansion in C9orf72 compromises nucleocytoplasmic transport. Nature. 2015;525(7567):129–33.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  55. Zhang K, Donnelly CJ, Haeusler AR, Grima JC, Machamer JB, Steinwald P, et al. The C9orf72 repeat expansion disrupts nucleocytoplasmic transport. Nature. 2015;525(7567):56–61.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  56. Maharjan N, Kunzli C, Buthey K, Saxena S. C9ORF72 regulates stress granule formation and its deficiency impairs stress granule assembly, hypersensitizing cells to stress. Mol Neurobiol. 2017;54(4):3062–77.

    Article  PubMed  CAS  Google Scholar 

  57. Schweizer Burguete A, Almeida S, Gao FB, Kalb R, Akins MR, Bonini NM. GGGGCC microsatellite RNA is neuritically localized, induces branching defects, and perturbs transport granule function. Elife. 2015;4:e08881.

    Article  Google Scholar 

  58. Mori K, Weng SM, Arzberger T, May S, Rentzsch K, Kremmer E, et al. The C9orf72 GGGGCC repeat is translated into aggregating dipeptide-repeat proteins in FTLD/ALS. Science. 2013;339(6125):1335–8.

    Article  CAS  PubMed  Google Scholar 

  59. Mizielinska S, Gronke S, Niccoli T, Ridler CE, Clayton EL, Devoy A, et al. C9orf72 repeat expansions cause neurodegeneration in Drosophila through arginine-rich proteins. Science. 2014;345(6201):1192–4.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  60. Schmidt EK, Clavarino G, Ceppi M, Pierre P. SUnSET, a nonradioactive method to monitor protein synthesis. Nat Methods. 2009;6(4):275–7.

    Article  PubMed  CAS  Google Scholar 

  61. Kanekura K, Yagi T, Cammack AJ, Mahadevan J, Kuroda M, Harms MB, et al. Poly-dipeptides encoded by the C9ORF72 repeats block global protein translation. Hum Mol Genet. 2016;25(9):1803–13.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  62. Kwon MS, Noh MY, Oh KW, Cho KA, Kang BY, Kim KS, et al. The immunomodulatory effects of human mesenchymal stem cells on peripheral blood mononuclear cells in ALS patients. J Neurochem. 2014;131(2):206–18.

    Article  PubMed  CAS  Google Scholar 

  63. Lee JE, Cooper TA. Pathogenic mechanisms of myotonic dystrophy. Biochem Soc Trans. 2009;37(Pt 6):1281–6.

    Article  PubMed  CAS  Google Scholar 

  64. Meola G, Cardani R. Myotonic dystrophy type 2 and modifier genes: an update on clinical and pathomolecular aspects. Neurol Sci. 2017;38(4):535–46.

    Article  PubMed  Google Scholar 

  65. Bates G. Huntingtin aggregation and toxicity in Huntington's disease. Lancet. 2003;361(9369):1642–4.

    Article  PubMed  CAS  Google Scholar 

  66. Savas JN, Ma B, Deinhardt K, Culver BP, Restituito S, Wu L, et al. A role for Huntington disease protein in dendritic RNA granules. J Biol Chem. 2010;285(17):13142–53.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  67. de Mezer M, Wojciechowska M, Napierala M, Sobczak K, Krzyzosiak WJ. Mutant CAG repeats of Huntingtin transcript fold into hairpins, form nuclear foci and are targets for RNA interference. Nucleic Acids Res. 2011;39(9):3852–63.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  68. Grima JC, Daigle JG, Arbez N, Cunningham KC, Zhang K, Ochaba J, et al. Mutant Huntingtin disrupts the nuclear pore complex. Neuron. 2017;94(1):93–107.e6.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  69. Gasset-Rosa F, Chillon-Marinas C, Goginashvili A, Atwal RS, Artates JW, Tabet R, et al. Polyglutamine-expanded Huntingtin exacerbates age-related disruption of nuclear integrity and nucleocytoplasmic transport. Neuron. 2017;94(1):48–57.e4.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  70. Fu YH, Kuhl DP, Pizzuti A, Pieretti M, Sutcliffe JS, Richards S, et al. Variation of the CGG repeat at the fragile X site results in genetic instability: resolution of the Sherman paradox. Cell. 1991;67(6):1047–58.

    Article  PubMed  CAS  Google Scholar 

  71. Kremer EJ, Pritchard M, Lynch M, Yu S, Holman K, Baker E, et al. Mapping of DNA instability at the fragile X to a trinucleotide repeat sequence p(CCG)n. Science. 1991;252(5013):1711–4.

    Article  CAS  PubMed  Google Scholar 

  72. Yu S, Pritchard M, Kremer E, Lynch M, Nancarrow J, Baker E, et al. Fragile X genotype characterized by an unstable region of DNA. Science. 1991;252(5010):1179–81.

    Article  PubMed  CAS  Google Scholar 

  73. Tassone F, Hagerman RJ, Chamberlain WD, Hagerman PJ. Transcription of the FMR1 gene in individuals with fragile X syndrome. Am J Med Genet. 2000;97(3):195–203.

    Article  PubMed  CAS  Google Scholar 

  74. Hagerman RJ, Leehey M, Heinrichs W, Tassone F, Wilson R, Hills J, et al. Intention tremor, parkinsonism, and generalized brain atrophy in male carriers of fragile X. Neurology. 2001;57(1):127–30.

    Article  PubMed  CAS  Google Scholar 

  75. Sofola OA, Jin P, Qin Y, Duan R, Liu H, de Haro M, et al. RNA-binding proteins hnRNP A2/B1 and CUGBP1 suppress fragile X CGG premutation repeat-induced neurodegeneration in a Drosophila model of FXTAS. Neuron. 2007;55(4):565–71.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  76. Jin P, Duan R, Qurashi A, Qin Y, Tian D, Rosser TC, et al. Pur alpha binds to rCGG repeats and modulates repeat-mediated neurodegeneration in a Drosophila model of fragile X tremor/ataxia syndrome. Neuron. 2007;55(4):556–64.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  77. Sellier C, Freyermuth F, Tabet R, Tran T, He F, Ruffenach F, et al. Sequestration of DROSHA and DGCR8 by expanded CGG RNA repeats alters microRNA processing in fragile X-associated tremor/ataxia syndrome. Cell Rep. 2013;3(3):869–80.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  78. Todd PK, Oh SY, Krans A, He F, Sellier C, Frazer M, et al. CGG repeat-associated translation mediates neurodegeneration in fragile X tremor ataxia syndrome. Neuron. 2013;78(3):440–55.

    Article  CAS  PubMed  Google Scholar 

  79. Oh SY, He F, Krans A, Frazer M, Taylor JP, Paulson HL, et al. RAN translation at CGG repeats induces ubiquitin proteasome system impairment in models of fragile X-associated tremor ataxia syndrome. Hum Mol Genet. 2015;24(15):4317–26.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  80. Sellier C, Buijsen RA, He F, Natla S, Jung L, Tropel P, et al. Translation of expanded CGG repeats into FMRpolyG is pathogenic and may contribute to fragile X tremor ataxia syndrome. Neuron. 2017;93(2):331–47.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  81. Maziuk B, Ballance HI, Wolozin B. Dysregulation of RNA binding protein aggregation in neurodegenerative disorders. Front Mol Neurosci. 2017;10:89.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  82. Iqbal K, Liu F, Gong CX, Grundke-Iqbal I. Tau in Alzheimer disease and related tauopathies. Curr Alzheimer Res. 2010;7(8):656–64.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  83. Vanderweyde T, Apicco DJ, Youmans-Kidder K, Ash PEA, Cook C, Lummertz da Rocha E, et al. Interaction of tau with the RNA-binding protein TIA1 regulates tau pathophysiology and toxicity. Cell Rep. 2016;15(7):1455–66.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  84. Daz-Muoz MDaKVYaNNLaCTaUJaTM. Tia1 dependent regulation of mRNA subcellular location and translation controls p53 expression in B cells. Nat Commun. 2017;8(1):530.

    Article  CAS  Google Scholar 

  85. Gu J, Wu F, Xu W, Shi J, Hu W, Jin N, et al. TDP-43 suppresses tau expression via promoting its mRNA instability. Nucleic Acids Res. 2017;45(10):6177–93.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  86. Ding Q, Markesbery WR, Chen Q, Li F, Keller JN. Ribosome dysfunction is an early event in Alzheimer’s disease. J Neurosci. 2005;25(40):9171–5.

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  87. Carthew RW, Sontheimer EJ. Origins and mechanisms of miRNAs and siRNAs. Cell. 2009;136(4):642–55.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  88. Kawahara Y, Mieda-Sato A. TDP-43 promotes microRNA biogenesis as a component of the Drosha and Dicer complexes. Proc Natl Acad Sci U S A. 2012;109(9):3347–52.

    Article  PubMed  PubMed Central  Google Scholar 

  89. Porta S, Kwong LK, Trojanowski JQ, Lee VM. Drosha inclusions are new components of dipeptide-repeat protein aggregates in FTLD-TDP and ALS C9orf72 expansion cases. J Neuropathol Exp Neurol. 2015;74(4):380–7.

    Article  PubMed  CAS  Google Scholar 

  90. Chou CC, Zhang Y, Umoh ME, Vaughan SW, Lorenzini I, Liu F, et al. TDP-43 pathology disrupts nuclear pore complexes and nucleocytoplasmic transport in 1 ALS/FTD. Nat Neurosci. 2018;21(2):228–39.

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  91. Eitan C, Hornstein E. Vulnerability of microRNA biogenesis in FTD-ALS. Brain Res. 2016;1647:105–11.

    Article  PubMed  CAS  Google Scholar 

  92. Gascon E, Lynch K, Ruan H, Almeida S, Verheyden JM, Seeley WW, et al. Alterations in microRNA-124 and AMPA receptors contribute to social behavioral deficits in frontotemporal dementia. Nat Med. 2014;20(12):1444–51.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  93. Zhang Z, Almeida S, Lu Y, Nishimura AL, Peng L, Sun D, et al. Downregulation of microRNA-9 in iPSC-derived neurons of FTD/ALS patients with TDP-43 mutations. PLoS One. 2013;8(10):e76055.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  94. Rinchetti P, Rizzuti M, Faravelli I, Corti S. MicroRNA metabolism and dysregulation in amyotrophic lateral sclerosis. Mol Neurobiol. 2018;55(3):2617–30.

    Article  PubMed  CAS  Google Scholar 

  95. Costa CJ, Willis DE. To the end of the line: axonal mRNA transport and local translation in health and neurodegenerative disease. Dev Neurobiol. 2018;78(3):209–20.

    Article  CAS  PubMed  Google Scholar 

  96. Spaulding EL, Burgess RW. Accumulating evidence for axonal translation in neuronal homeostasis. Front Neurosci. 2017;11:312.

    Article  PubMed  PubMed Central  Google Scholar 

  97. Cestra G, Rossi S, Di Salvio M, Cozzolino M. Control of mRNA translation in ALS proteinopathy. Front Mol Neurosci. 2017;10:85.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  98. Shi Z, Fujii K, Kovary KM, Genuth NR, Rost HL, Teruel MN, et al. Heterogeneous ribosomes preferentially translate distinct subpools of mRNAs genome-wide. Mol Cell. 2017;67(1):71–83.e7.

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  99. Ernst V, Levin DH, London IM. In situ phosphorylation of the alpha subunit of eukaryotic initiation factor 2 in reticulocyte lysates inhibited by heme deficiency, double-stranded RNA, oxidized glutathione, or the heme-regulated protein kinase. Proc Natl Acad Sci U S A. 1979;76(5):2118–22.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  100. Moreno JA, Halliday M, Molloy C, Radford H, Verity N, Axten JM, et al. Oral treatment targeting the unfolded protein response prevents neurodegeneration and clinical disease in prion-infected mice. Sci Transl Med. 2013;5(206):206ra138.

    Article  PubMed  CAS  Google Scholar 

  101. Briggs DI, Defensor E, Memar Ardestani P, Yi B, Halpain M, Seabrook G, et al. Role of endoplasmic reticulum stress in learning and memory impairment and Alzheimer’s disease-like neuropathology in the PS19 and APPSwe mouse models of tauopathy and amyloidosis. eNeuro. 2017;4(4). https://doi.org/10.1523/ENEURO.0025-17.2017.

    Article  PubMed  PubMed Central  Google Scholar 

  102. Johnson EC, Kang J. A small molecule targeting protein translation does not rescue spatial learning and memory deficits in the hAPP-J20 mouse model of Alzheimer’s disease. PeerJ. 2016;4:e2565.

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Daniela C. Zarnescu .

Editor information

Editors and Affiliations

Rights and permissions

Reprints and permissions

Copyright information

© 2018 Springer International Publishing AG, part of Springer Nature

About this chapter

Check for updates. Verify currency and authenticity via CrossMark

Cite this chapter

Lehmkuhl, E.M., Zarnescu, D.C. (2018). Lost in Translation: Evidence for Protein Synthesis Deficits in ALS/FTD and Related Neurodegenerative Diseases. In: Sattler, R., Donnelly, C. (eds) RNA Metabolism in Neurodegenerative Diseases. Advances in Neurobiology, vol 20. Springer, Cham. https://doi.org/10.1007/978-3-319-89689-2_11

Download citation

Publish with us

Policies and ethics