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.

  • Original Article
  • Published:

Transcriptome profiling of UPF3B/NMD-deficient lymphoblastoid cells from patients with various forms of intellectual disability

Abstract

The nonsense-mediated mRNA decay (NMD) pathway was originally discovered by virtue of its ability to rapidly degrade aberrant mRNAs with premature termination codons. More recently, it was shown that NMD also directly regulates subsets of normal transcripts, suggesting that NMD has roles in normal biological processes. Indeed, several NMD factors have been shown to regulate neurological events (for example, neurogenesis and synaptic plasticity) in numerous vertebrate species. In man, mutations in the NMD factor gene UPF3B, which disrupts a branch of the NMD pathway, cause various forms of intellectual disability (ID). Using Epstein Barr virus—immortalized B cells, also known as lymphoblastoid cell lines (LCLs), from ID patients that have loss-of-function mutations in UPF3B, we investigated the genome-wide consequences of compromised NMD and the role of NMD in neuronal development and function. We found that 5% of the human transcriptome is impacted in UPF3B patients. The UPF3B paralog, UPF3A, is stabilized in all UPF3B patients, and partially compensates for the loss of UPF3B function. Interestingly, UPF3A protein, but not mRNA, was stabilised in a quantitative manner that inversely correlated with the severity of patients' phenotype. This suggested that the ability to stabilize the UPF3A protein is a crucial modifier of the neurological symptoms due to loss of UPF3B. We also identified ARHGAP24, which encodes a GTPase-activating protein, as a canonical target of NMD, and we provide evidence that deregulation of this gene inhibits axon and dendrite outgrowth and branching. Our results demonstrate that the UPF3B-dependent NMD pathway is a major regulator of the transcriptome and that its targets have important roles in neuronal cells.

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
Figure 2
Figure 3

Similar content being viewed by others

Accession codes

Accessions

GenBank/EMBL/DDBJ

References

  1. Nicholson P, Yepiskoposyan H, Metze S, Zamudio Orozco R, Kleinschmidt N, Muhlemann O . Nonsense-mediated mRNA decay in human cells: mechanistic insights, functions beyond quality control and the double-life of NMD factors. Cell Mol Life Sci 2010; 67: 677–700.

    Article  CAS  Google Scholar 

  2. Gehring NH, Neu-Yilik G, Schell T, Hentze MW, Kulozik AE . Y14 and hUpf3b form an NMD-activating complex. Mol Cell 2003; 11: 939–949.

    Article  CAS  Google Scholar 

  3. Kim VN, Kataoka N, Dreyfuss G . Role of the nonsense-mediated decay factor hUpf3 in the splicing-dependent exon-exon junction complex. Science 2001; 293: 1832–1836.

    Article  CAS  Google Scholar 

  4. 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 2008; 15: 85–93.

    Article  CAS  Google Scholar 

  5. Kashima I, Yamashita A, Izumi N, Kataoka N, Morishita R, Hoshino S 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 2006; 20: 355–367.

    Article  CAS  Google Scholar 

  6. Ivanov PV, Gehring NH, Kunz JB, Hentze MW, Kulozik AE . Interactions between UPF1, eRFs, PABP and the exon junction complex suggest an integrated model for mammalian NMD pathways. EMBO J 2008; 27: 736–747.

    Article  CAS  Google Scholar 

  7. Singh G, Rebbapragada I, Lykke-Andersen J . A competition between stimulators and antagonists of Upf complex recruitment governs human nonsense-mediated mRNA decay. PLoS Biol 2008; 6: e111.

    Article  Google Scholar 

  8. Gehring NH, Kunz JB, Neu-Yilik G, Breit S, Viegas MH, Hentze MW et al. Exon-junction complex components specify distinct routes of nonsense-mediated mRNA decay with differential cofactor requirements. Mol Cell 2005; 20: 65–75.

    Article  CAS  Google Scholar 

  9. Chan WK, Huang L, Gudikote JP, Chang YF, Imam JS, MacLean II JA et al. An alternative branch of the nonsense-mediated decay pathway. EMBO J 2007; 26: 1820–1830.

    Article  CAS  Google Scholar 

  10. Chan WK, Bhalla AD, Le Hir H, Nguyen LS, Huang L, Gecz J et al. A UPF3-mediated regulatory switch that maintains RNA surveillance. Nat Struct Mol Biol 2009; 16: 747–753.

    Article  CAS  Google Scholar 

  11. Kunz JB, Neu-Yilik G, Hentze MW, Kulozik AE, Gehring NH . Functions of hUpf3a and hUpf3b in nonsense-mediated mRNA decay and translation. RNA 2006; 12: 1015–1022.

    Article  CAS  Google Scholar 

  12. Mendell JT, Sharifi NA, Meyers JL, Martinez-Murillo F, Dietz HC . Nonsense surveillance regulates expression of diverse classes of mammalian transcripts and mutes genomic noise. Nat Genet 2004; 36: 1073–1078.

    Article  CAS  Google Scholar 

  13. Wittmann J, Hol EM, Jack HM . hUPF2 silencing identifies physiologic substrates of mammalian nonsense-mediated mRNA decay. Mol Cell Biol 2006; 26: 1272–1287.

    Article  CAS  Google Scholar 

  14. Weischenfeldt J, Damgaard I, Bryder D, Theilgaard-Monch K, Thoren LA, Nielsen FC et al. NMD is essential for hematopoietic stem and progenitor cells and for eliminating by-products of programmed DNA rearrangements. Genes Dev 2008; 22: 1381–1396.

    Article  CAS  Google Scholar 

  15. Rehwinkel J, Letunic I, Raes J, Bork P, Izaurralde E . Nonsense-mediated mRNA decay factors act in concert to regulate common mRNA targets. RNA 2005; 11: 1530–1544.

    Article  CAS  Google Scholar 

  16. He F, Li X, Spatrick P, Casillo R, Dong S, Jacobson A . Genome-wide analysis of mRNAs regulated by the nonsense-mediated and 5′ to 3′ mRNA decay pathways in yeast. Mol Cell 2003; 12: 1439–1452.

    Article  CAS  Google Scholar 

  17. Medghalchi SM, Frischmeyer PA, Mendell JT, Kelly AG, Lawler AM, Dietz HC . Rent1, a trans-effector of nonsense-mediated mRNA decay, is essential for mammalian embryonic viability. Hum Mol Genet 2001; 10: 99–105.

    Article  CAS  Google Scholar 

  18. Laumonnier F, Shoubridge C, Antar C, Nguyen LS, Van Esch H, Kleefstra T 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 2010; 15: 767–776.

    Article  CAS  Google Scholar 

  19. Nguyen LS, Tarpey PS, Raymond FL, Rodriguez J, Hackett A, Vandeleur L et al. Mutations in UPF3B, a member of the nonsense-mediated mRNA decay complex, cause syndromic and nonsyndromic mental retardation. Nat Genet 2007; 39: 1127–1133.

    Article  Google Scholar 

  20. Addington AM, Gauthier J, Piton A, Hamdan FF, Raymond A, Gogtay N et al. A novel frameshift mutation in UPF3B identified in brothers affected with childhood onset schizophrenia and autism spectrum disorders. Mol Psychiatry 2011; 16: 238–239.

    Article  CAS  Google Scholar 

  21. Mortazavi A, Williams BA, McCue K, Schaeffer L, Wold B . Mapping and quantifying mammalian transcriptomes by RNA-Seq. Nat Methods 2008; 5: 621–628.

    Article  CAS  Google Scholar 

  22. Tang F, Barbacioru C, Wang Y, Nordman E, Lee C, Xu N et al. mRNA-Seq whole-transcriptome analysis of a single cell. Nat Methods 2009; 6: 377–382.

    Article  CAS  Google Scholar 

  23. Irizarry RA, Hobbs B, Collin F, Beazer-Barclay YD, Antonellis KJ, Scherf U et al. Exploration, normalization, and summaries of high density oligonucleotide array probe level data. Biostatistics 2003; 4: 249–264.

    Article  Google Scholar 

  24. Diskin SJ, Li M, Hou C, Yang S, Glessner J, Hakonarson H et al. Adjustment of genomic waves in signal intensities from whole-genome SNP genotyping platforms. Nucleic Acids Res 2008; 36: e126.

    Article  Google Scholar 

  25. Olshen AB, Venkatraman ES, Lucito R, Wigler M . Circular binary segmentation for the analysis of array-based DNA copy number data. Biostatistics 2004; 5: 557–572.

    Article  Google Scholar 

  26. McLaren W, Pritchard B, Rios D, Chen Y, Flicek P, Cunningham F . Deriving the consequences of genomic variants with the Ensembl API and SNP Effect Predictor. Bioinformatics 2010; 26: 2069–2070.

    Article  CAS  Google Scholar 

  27. Kumar P, Henikoff S, Ng PC . Predicting the effects of coding non-synonymous variants on protein function using the SIFT algorithm. Nat Protoc 2009; 4: 1073–1081.

    Article  CAS  Google Scholar 

  28. Adzhubei IA, Schmidt S, Peshkin L, Ramensky VE, Gerasimova A, Bork P et al. A method and server for predicting damaging missense mutations. Nat Methods 2010; 7: 248–249.

    Article  CAS  Google Scholar 

  29. Su AI, Wiltshire T, Batalov S, Lapp H, Ching KA, Block D et al. A gene atlas of the mouse and human protein-encoding transcriptomes. Proc Natl Acad Sci USA 2004; 101: 6062–6067.

    Article  CAS  Google Scholar 

  30. Kaech S, Banker G . Culturing hippocampal neurons. Nat Protoc 2006; 1: 2406–2415.

    Article  CAS  Google Scholar 

  31. Usuki F, Yamashita A, Kashima I, Higuchi I, Osame M, Ohno S . Specific inhibition of nonsense-mediated mRNA decay components, SMG-1 or Upf1, rescues the phenotype of Ullrich disease fibroblasts. Mol Ther 2006; 14: 351–360.

    Article  CAS  Google Scholar 

  32. Miyagishi M, Taira K . U6 promoter-driven siRNAs with four uridine 3′ overhangs efficiently suppress targeted gene expression in mammalian cells. Nat Biotechnol 2002; 20: 497–500.

    Article  CAS  Google Scholar 

  33. Wang ET, Sandberg R, Luo S, Khrebtukova I, Zhang L, Mayr C et al. Alternative isoform regulation in human tissue transcriptomes. Nature 2008; 456: 470–476.

    Article  CAS  Google Scholar 

  34. Rehwinkel J, Raes J, Izaurralde E . Nonsense-mediated mRNA decay: Target genes and functional diversification of effectors. Trends Biochem Sci 2006; 31: 639–646.

    Article  CAS  Google Scholar 

  35. Ramakers GJ . Rho proteins, mental retardation and the cellular basis of cognition. Trends Neurosci 2002; 25: 191–199.

    Article  CAS  Google Scholar 

  36. Fox JW, Lamperti ED, Eksioglu YZ, Hong SE, Feng Y, Graham DA et al. Mutations in filamin 1 prevent migration of cerebral cortical neurons in human periventricular heterotopia. Neuron 1998; 21: 1315–1325.

    Article  CAS  Google Scholar 

  37. Ohta Y, Hartwig JH, Stossel TP . FilGAP, a Rho- and ROCK-regulated GAP for Rac binds filamin A to control actin remodelling. Nat Cell Biol 2006; 8: 803–814.

    Article  CAS  Google Scholar 

  38. Lavelin I, Geiger B . Characterization of a novel GTPase-activating protein associated with focal adhesions and the actin cytoskeleton. J Biol Chem 2005; 280: 7178–7185.

    Article  CAS  Google Scholar 

  39. Carter MS, Doskow J, Morris P, Li S, Nhim RP, Sandstedt 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 1995; 270: 28995–29003.

    Article  CAS  Google Scholar 

  40. Silver DL, Watkins-Chow DE, Schreck KC, Pierfelice TJ, Larson DM, Burnetti AJ et al. The exon junction complex component Magoh controls brain size by regulating neural stem cell division. Nat Neurosci 2010; 13: 551–558.

    Article  CAS  Google Scholar 

  41. Giorgi C, Yeo GW, Stone ME, Katz DB, Burge C, Turrigiano G et al. The EJC factor eIF4AIII modulates synaptic strength and neuronal protein expression. Cell 2007; 130: 179–191.

    Article  CAS  Google Scholar 

  42. Long AA, Mahapatra CT, Woodruff III EA, Rohrbough J, Leung HT, Shino S et al. The nonsense-mediated decay pathway maintains synapse architecture and synaptic vesicle cycle efficacy. J Cell Sci 2010; 123 (Part 19): 3303–3315.

    Article  CAS  Google Scholar 

  43. Bruno IG, Karam R, Huang L, Bhardwaj A, Lou CH, Shum EY et al. Identification of a MicroRNA that Activates Gene Expression by Repressing Nonsense-Mediated RNA Decay. Mol Cell 2011; 42: 500–510.

    Article  CAS  Google Scholar 

  44. Machacek M, Hodgson L, Welch C, Elliott H, Pertz O, Nalbant P et al. Coordination of Rho GTPase activities during cell protrusion. Nature 2009; 461: 99–103.

    Article  CAS  Google Scholar 

  45. Nakamura F, Heikkinen O, Pentikainen OT, Osborn TM, Kasza KE, Weitz DA et al. Molecular basis of filamin A-FilGAP interaction and its impairment in congenital disorders associated with filamin A mutations. PLoS One 2009; 4: e4928.

    Article  Google Scholar 

  46. Durand CM, Perroy J, Loll F, Perrais D, Fagni L, Bourgeron T et al. SHANK3 mutations identified in autism lead to modification of dendritic spine morphology via an actin-dependent mechanism. Mol Psychiatry 2011 (e-pub ahead of print).

  47. Hoogenraad CC, Koekkoek B, Akhmanova A, Krugers H, Dortland B, Miedema M et al. Targeted mutation of Cyln2 in the Williams syndrome critical region links CLIP-115 haploinsufficiency to neurodevelopmental abnormalities in mice. Nat Genet 2002; 32: 116–127.

    Article  CAS  Google Scholar 

  48. Kobayashi M, Toyama R, Takeda H, Dawid IB, Kawakami K . Overexpression of the forebrain-specific homeobox gene six3 induces rostral forebrain enlargement in zebrafish. Development 1998; 125: 2973–2982.

    CAS  PubMed  Google Scholar 

  49. Lagutin OV, Zhu CC, Kobayashi D, Topczewski J, Shimamura K, Puelles L et al. Six3 repression of Wnt signaling in the anterior neuroectoderm is essential for vertebrate forebrain development. Genes Dev 2003; 17: 368–379.

    Article  CAS  Google Scholar 

  50. Wallis DE, Roessler E, Hehr U, Nanni L, Wiltshire T, Richieri-Costa A et al. Mutations in the homeodomain of the human SIX3 gene cause holoprosencephaly. Nat Genet 1999; 22: 196–198.

    Article  CAS  Google Scholar 

  51. Appolloni I, Calzolari F, Corte G, Perris R, Malatesta P . Six3 controls the neural progenitor status in the murine CNS. Cereb Cortex 2008; 18: 553–562.

    Article  Google Scholar 

  52. Doe CM, Relkovic D, Garfield AS, Dalley JW, Theobald DE, Humby T et al. Loss of the imprinted snoRNA mbii-52 leads to increased 5htr2c pre-RNA editing and altered 5HT2CR-mediated behaviour. Hum Mol Genet 2009; 18: 2140–2148.

    Article  CAS  Google Scholar 

  53. Kishore S, Stamm S . Regulation of alternative splicing by snoRNAs. Cold Spring Harb Symp Quant Biol 2006; 71: 329–334.

    Article  CAS  Google Scholar 

  54. Sahoo T, del Gaudio D, German JR, Shinawi M, Peters SU, Person RE et al. Prader-Willi phenotype caused by paternal deficiency for the HBII-85 C/D box small nucleolar RNA cluster. Nat Genet 2008; 40: 719–721.

    Article  CAS  Google Scholar 

  55. Chamberlain SJ, Lalande M . Neurodevelopmental disorders involving genomic imprinting at human chromosome 15q11-q13. Neurobiol Dis 2010; 39: 13–20.

    Article  CAS  Google Scholar 

  56. Runte M, Kroisel PM, Gillessen-Kaesbach G, Varon R, Horn D, Cohen MY et al. SNURF-SNRPN and UBE3A transcript levels in patients with Angelman syndrome. Hum Genet 2004; 114: 553–561.

    Article  CAS  Google Scholar 

  57. Ramocki MB, Zoghbi HY . Failure of neuronal homeostasis results in common neuropsychiatric phenotypes. Nature 2008; 455: 912–918.

    Article  CAS  Google Scholar 

  58. Shi L, Jones WD, Jensen RV, Harris SC, Perkins RG, Goodsaid FM et al. The balance of reproducibility, sensitivity, and specificity of lists of differentially expressed genes in microarray studies. BMC Bioinformatics 2008; 9 (Suppl 9): S10.

    Article  Google Scholar 

  59. Lelivelt MJ, Culbertson MR . Yeast Upf proteins required for RNA surveillance affect global expression of the yeast transcriptome. Mol Cell Biol 1999; 19: 6710–6719.

    Article  CAS  Google Scholar 

  60. Khajavi M, Inoue K, Lupski JR . Nonsense-mediated mRNA decay modulates clinical outcome of genetic disease. Eur J Hum Genet 2006; 14: 1074–1081.

    Article  CAS  Google Scholar 

  61. Bevilacqua L, Doly S, Kaprio J, Yuan Q, Tikkanen R, Paunio T et al. A population-specific HTR2B stop codon predisposes to severe impulsivity. Nature 2010; 468: 1061–1066.

    Article  CAS  Google Scholar 

  62. Dixon AL, Liang L, Moffatt MF, Chen W, Heath S, Wong KC et al. A genome-wide association study of global gene expression. Nat Genet 2007; 39: 1202–1207.

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank the members of the families studied for participation and Brigitte Jauffrion and Lucianne Vandeleur for help with cell culture. This work was supported by grants from the Australian NH&MRC Project Grant 453457 to JoG, and the Women's and Children's Hospital Foundation Grant to LJ and JoG. JoG is supported by NH&MRC Principal Research Fellowship. LSN's PhD scholarship was supported by the MS McLeod Foundation.

Author contributions: LSN performed most of the data analysis; LJ performed the overexpression of ARHGAP24 in mouse primary hippocampal neurons; CS performed the overexpression of ARHGAP24 in mouse PC12 cell line; WC, LH and MFW contributed the microarray and reverse-transcriptase quantitative PCR data of NMD knockdown in HeLa cells; LH performed the RNAi knockdown of UPF1, UPF2 and SMG1 in HeLa cells; SS performed the in situ hybridization of ARHGAP24 in human brain sample; CH and JeG generated the antibody against ARHGAP24 isoform 1 and the ARHGAP24 isoform 1 clone; FL, MR, AH, MF, JR, AKS, LY, AAM and JLR contributed with patient material or LCL lines for this work; MC provided advice on the data analysis; This project was designed and supervised by JoG; The paper was written by LSN, LJ and JoG, with advice from MFW.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to J Gecz.

Ethics declarations

Competing interests

The authors declare no conflict of interest.

Additional information

Supplementary Information accompanies the paper on the Molecular Psychiatry website

Supplementary information

PowerPoint slides

Rights and permissions

Reprints and permissions

About this article

Cite this article

Nguyen, L., Jolly, L., Shoubridge, C. et al. Transcriptome profiling of UPF3B/NMD-deficient lymphoblastoid cells from patients with various forms of intellectual disability. Mol Psychiatry 17, 1103–1115 (2012). https://doi.org/10.1038/mp.2011.163

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/mp.2011.163

Keywords

This article is cited by

Search

Quick links