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.

  • Analysis
  • Published:

Integrative transcriptome analyses of the aging brain implicate altered splicing in Alzheimer’s disease susceptibility

Abstract

Here we use deep sequencing to identify sources of variation in mRNA splicing in the dorsolateral prefrontal cortex (DLPFC) of 450 subjects from two aging cohorts. Hundreds of aberrant pre-mRNA splicing events are reproducibly associated with Alzheimer’s disease. We also generate a catalog of splicing quantitative trait loci (sQTL) effects: splicing of 3,006 genes is influenced by genetic variation. We report that altered splicing is the mechanism for the effects of the PICALM, CLU and PTK2B susceptibility alleles. Furthermore, we performed a transcriptome-wide association study and identified 21 genes with significant associations with Alzheimer’s disease, many of which are found in known loci, whereas 8 are in novel loci. These results highlight the convergence of old and new genes associated with Alzheimer’s disease in autophagy–lysosomal-related pathways. Overall, this study of the transcriptome of the aging brain provides evidence that dysregulation of mRNA splicing is a feature of Alzheimer’s disease and is, in some cases, genetically driven.

This is a preview of subscription content, access via your institution

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: Overview of the study.
Fig. 2: Differential splicing analysis in relation to Alzheimer’s disease diagnosis and neuropathology.
Fig. 3: Enrichment of sQTLs in epigenomic marks and in Alzheimer’s disease GWAS.
Fig. 4: Enrichment of RBP binding sites among sQTLs.
Fig. 5: TWAS of Alzheimer’s disease.
Fig. 6: TWAS prioritizes Alzheimer’s disease genes in endocytosis- and autophagy-related pathways.

Similar content being viewed by others

Data availability

The ROS/MAP sQTL visualization (Shiny App) browser is available at https://rajlab.shinyapps.io/sQTLviz_ROSMAP/. The ROS/MAP data are available at the RADC Research Resource Sharing Hub at http://www.radc.rush.edu/. The ROS/MAP and MSBB mapped RNA-seq data that support the findings of this study are available from the AMP-AD Knowledge Portal (https://www.synapse.org/#!Synapse:syn2580853) upon authentication by the Consortium. The CMC data are available from the CMC Knowledge Portal (https://www.synapse.org/#!Synapse:syn4923029).

References

  1. Wang, E. T. et al. Alternative isoform regulation in human tissue transcriptomes. Nature 456, 470–476 (2008).

    Article  CAS  Google Scholar 

  2. Kornblihtt, A. R. et al. Alternative splicing: a pivotal step between eukaryotic transcription and translation. Nat. Rev. Mol. Cell Biol. 14, 153–165 (2013).

    Article  CAS  Google Scholar 

  3. Barbosa-Morais, N. L. et al. The evolutionary landscape of alternative splicing in vertebrate species. Science 338, 1587–1593 (2012).

    Article  CAS  Google Scholar 

  4. Wang, G. S. & Cooper, T. A. Splicing in disease: disruption of the splicing code and the decoding machinery. Nat. Rev. Genet. 8, 749–761 (2007).

    Article  CAS  Google Scholar 

  5. Dredge, B. K., Polydorides, A. D. & Darnell, R. B. The splice of life: alternative splicing and neurological disease. Nat. Rev. Neurosci. 2, 43–50 (2001).

    Article  CAS  Google Scholar 

  6. Parikshak, N. N. et al. Genome-wide changes in lncRNA, splicing, and regional gene expression patterns in autism. Nature 540, 423–427 (2016).

    Article  CAS  Google Scholar 

  7. Arai, T. et al. TDP-43 is a component of ubiquitin-positive tau-negative inclusions in frontotemporal lobar degeneration and amyotrophic lateral sclerosis. Biochem. Biophys. Res. Commun. 351, 602–611 (2006).

    Article  CAS  Google Scholar 

  8. Trabzuni, D. et al. MAPT expression and splicing is differentially regulated by brain region: relation to genotype and implication for tauopathies. Hum. Mol. Genet. 21, 4094–4103 (2012).

    Article  CAS  Google Scholar 

  9. Rockenstein, E. M. et al. Levels and alternative splicing of amyloid beta protein precursor (APP) transcripts in brains of APP transgenic mice and humans with Alzheimer’s disease. J. Biol. Chem. 270, 28257–28267 (1995).

    Article  CAS  Google Scholar 

  10. Buee, L., Bussiere, T., Buee-Scherrer, V., Delacourte, A. & Hof, P. R. Tau protein isoforms, phosphorylation and role in neurodegenerative disorders. Brain Res. Rev. 33, 95–130 (2000).

    Article  CAS  Google Scholar 

  11. Valenca, G. T. et al. The role of MAPT haplotype H2 and isoform 1N/4R in Parkinsonism of older adults. PLoS ONE 11, e0157452 (2016).

    Article  Google Scholar 

  12. Bai, B. et al. U1 small nuclear ribonucleoprotein complex and RNA splicing alterations in Alzheimer’s disease. Proc. Natl Acad. Sci. USA 110, 16562–16567 (2013).

    Article  CAS  Google Scholar 

  13. Vaquero-Garcia, J. et al. A new view of transcriptome complexity and regulation through the lens of local splicing variations. eLife 5, e11752 (2016).

    Article  Google Scholar 

  14. Lambert, J. C. et al. Meta-analysis of 74,046 individuals identifies 11 new susceptibility loci for Alzheimer’s disease. Nat. Genet. 45, 1452–1458 (2013).

    Article  CAS  Google Scholar 

  15. Raj, T. et al. Polarization of the effects of autoimmune and neurodegenerative risk alleles in leukocytes. Science 344, 519–523 (2014).

    Article  CAS  Google Scholar 

  16. Raj, T. et al. CD33: increased inclusion of exon 2 implicates the Ig V-set domain in Alzheimer’s disease susceptibility. Hum. Mol. Genet. 23, 2729–2736 (2014).

    Article  CAS  Google Scholar 

  17. Bennett, D. A., Schneider, J. A., Arvanitakis, Z. & Wilson, R. S. Overview and findings from the Religious Orders Study. Curr. Alzheimer. Res. 9, 628–645 (2012).

    Article  CAS  Google Scholar 

  18. Bennett, D. A. et al. Selected findings from the Religious Orders Study and Rush Memory and Aging Project. J. Alzheimers. Dis. 33, S397–S403 (2013).

    Article  Google Scholar 

  19. Mostafavi, S. et al. A molecular network of the aging human brain provides insights into the pathology and cognitive decline of Alzheimer’s disease. Nat. Neurosci. 21, 811–819 (2018).

    Article  CAS  Google Scholar 

  20. Li, Y. I. et al. Annotation-free quantification of RNA splicing using LeafCutter. Nat. Genet. 50, 151–158 (2018).

    Article  CAS  Google Scholar 

  21. Li, Y. I. et al. RNA splicing is a primary link between genetic variation and disease. Science 352, 600–604 (2016).

    Article  CAS  Google Scholar 

  22. Tollervey, J. R. et al. Analysis of alternative splicing associated with aging and neurodegeneration in the human brain. Genome Res. 21, 1572–1582 (2011).

    Article  CAS  Google Scholar 

  23. Mitchelmore, C. et al. NDRG2: a novel Alzheimer’s disease associated protein. Neurobiol. Dis. 16, 48–58 (2004).

    Article  CAS  Google Scholar 

  24. Zhang, B. & Horvath, S. A general framework for weighted gene co-expression network analysis. Stat. Appl. Genet. Mol. Biol. 4, e17 (2005).

    Article  Google Scholar 

  25. Wang, M. et al. Integrative network analysis of nineteen brain regions identifies molecular signatures and networks underlying selective regional vulnerability to Alzheimer’s disease. Genome Med. 8, 104 (2016).

    Article  CAS  Google Scholar 

  26. Ongen, H., Buil, A., Brown, A. A., Dermitzakis, E. T. & Delaneau, O. Fast and efficient QTL mapper for thousands of molecular phenotypes. Bioinformatics 32, 1479–1485 (2016).

    Article  CAS  Google Scholar 

  27. Bernstein, B. E. et al. The NIH Roadmap Epigenomics Mapping Consortium. Nat. Biotechnol. 28, 1045–1048 (2010).

    Article  CAS  Google Scholar 

  28. Fromer, M. et al. Gene expression elucidates functional impact of polygenic risk for schizophrenia. Nat. Neurosci. 19, 1442–1453 (2016).

    Article  CAS  Google Scholar 

  29. Ng, B. et al. An xQTL map integrates the genetic architecture of the human brain’s transcriptome and epigenome. Nat. Neurosci. 20, 1418–1426 (2017).

    Article  CAS  Google Scholar 

  30. Nicolae, D. L. et al. Trait-associated SNPs are more likely to be eQTLs: annotation to enhance discovery from GWAS. PLoS Genet. 6, e1000888 (2010).

    Article  Google Scholar 

  31. Chen, L. et al. Genetic drivers of epigenetic and transcriptional variation in human immune cells. Cell 167, 1398–1414 (2016).

    Article  CAS  Google Scholar 

  32. Malik, M. et al. CD33 Alzheimer’s risk-altering polymorphism, CD33 expression, and exon 2 splicing. J. Neurosci. 33, 13320–13325 (2013).

    Article  CAS  Google Scholar 

  33. Sibley, C. R., Blazquez, L. & Ule, J. Lessons from non-canonical splicing. Nat. Rev. Genet. 17, 407–421 (2016).

    Article  CAS  Google Scholar 

  34. Yang, Y. C. et al. CLIPdb: a CLIP-seq database for protein–RNA interactions. BMC Genomics 16, 51 (2015).

    Article  CAS  Google Scholar 

  35. Scheckel, C. et al. Regulatory consequences of neuronal ELAV-like protein binding to coding and non-coding RNAs in human brain. eLife 5, e10421 (2016).

    Article  Google Scholar 

  36. Borreca, A., Gironi, K., Amadoro, G. & Ammassari-Teule, M. Opposite dysregulation of fragile-X mental retardation protein and heteronuclear ribonucleoprotein C protein associates with enhanced APP translation in Alzheimer disease. Mol. Neurobiol. 53, 3227–3234 (2016).

    Article  CAS  Google Scholar 

  37. Gusev, A. et al. Integrative approaches for large-scale transcriptome-wide association studies. Nat. Genet. 48, 245–252 (2016).

    Article  CAS  Google Scholar 

  38. Seyfried, N. T. et al. A multi-network approach identifies protein-specific co-expression in asymptomatic and symptomatic Alzheimer’s disease. Cell Syst. 4, 60–72 (2017).

    Article  CAS  Google Scholar 

  39. Liu, J. Z., Erlich, Y. & Pickrell, J. K. Case–control association mapping by proxy using family history of disease. Nat. Genet. 49, 325–331 (2017).

    Article  CAS  Google Scholar 

  40. Gusev, A. et al. Transcriptome-wide association study of schizophrenia and chromatin activity yields mechanistic disease insights. Nat. Genet. 50, 538–548 (2018).

    Article  CAS  Google Scholar 

  41. Yang, J. et al. Conditional and joint multiple-SNP analysis of GWAS summary statistics identifies additional variants influencing complex traits. Nat. Genet. 44, 369–375 (2012).

    Article  CAS  Google Scholar 

  42. Li, T. et al. GeNets: a unified web platform for network-based genomic analyses. Nat. Methods 15, 543–546 (2018).

    Article  CAS  Google Scholar 

  43. Raj, T. et al. Alzheimer disease susceptibility loci: evidence for a protein network under natural selection. Am. J. Hum. Genet. 90, 720–726 (2012).

    Article  CAS  Google Scholar 

  44. Nixon, R. A. New perspectives on lysosomes in ageing and neurodegenerative disease. Ageing Res. Rev. 32, 1 (2016).

    Article  CAS  Google Scholar 

  45. Emmett, M. J. et al. Histone deacetylase 3 prepares brown adipose tissue for acute thermogenic challenge. Nature 546, 544–548 (2017).

    Article  CAS  Google Scholar 

  46. Tian, Y., Chang, J. C., Fan, E. Y., Flajolet, M. & Greengard, P. Adaptor complex AP2/PICALM, through interaction with LC3, targets Alzheimer’s APP-CTF for terminal degradation via autophagy. Proc. Natl Acad. Sci. USA 110, 17071–17076 (2013).

    Article  CAS  Google Scholar 

  47. Ingelsson, M. et al. Early Aβ accumulation and progressive synaptic loss, gliosis, and tangle formation in AD brain. Neurology 62, 925–931 (2004).

    Article  CAS  Google Scholar 

  48. Guillozet, A. L., Weintraub, S., Mash, D. C. & Mesulam, M. M. Neurofibrillary tangles, amyloid, and memory in aging and mild cognitive impairment. Arch. Neurol. 60, 729–736 (2003).

    Article  Google Scholar 

  49. The GTEx Consortium. The Genotype-Tissue Expression (GTEx) pilot analysis: multitissue gene regulation in humans. Science 348, 648–660 (2015).

    Article  Google Scholar 

  50. Fairfax, B. P. et al. Innate immune activity conditions the effect of regulatory variants upon monocyte gene expression. Science 343, 1246949 (2014).

    Article  Google Scholar 

  51. Bennett, D. A. et al. Overview and findings from the Rush Memory and Aging Project. Curr. Alzheimer. Res. 9, 646–663 (2012).

    Article  CAS  Google Scholar 

  52. Purcell, S. et al. PLINK: a tool set for whole-genome association and population-based linkage analyses. Am. J. Hum. Genet. 81, 559–575 (2007).

    Article  CAS  Google Scholar 

  53. Patterson, N., Price, A. L. & Reich, D. Population structure and Eigenanalysis. PLoS Genet. 2, e190 (2006).

    Article  Google Scholar 

  54. Das, S. et al. Next-generation genotype imputation service and methods. Nat. Genet. 48, 1284–1287 (2016).

    Article  CAS  Google Scholar 

  55. The Haplotype Reference Consortium. A reference panel of 64,976 haplotypes for genotype imputation. Nat. Genet. 48, 1279–1283 (2016).

    Article  Google Scholar 

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

    Article  CAS  Google Scholar 

  57. Li, B. & Dewey, C. N. RSEM: accurate transcript quantification from RNA-seq data with or without a reference genome. BMC Bioinformatics 12, 323 (2011).

    Article  CAS  Google Scholar 

  58. Johnson, W. E., Li, C. & Rabinovic, A. Adjusting batch effects in microarray expression data using empirical Bayes methods. Biostatistics 8, 118–127 (2007).

    Article  Google Scholar 

  59. Hoffman, G. E. & Schadt, E. E. variancePartition: interpreting drivers of variation in complex gene expression studies. BMC Bioinformatics 17, 483 (2016).

    Article  Google Scholar 

  60. van de Geijn, B., McVicker, G., Gilad, Y. & Pritchard, J. K. WASP: allele-specific software for robust molecular quantitative trait locus discovery. Nat. Methods 12, 1061–1063 (2015).

    Article  Google Scholar 

  61. Degner, J. F. et al. DNase I sensitivity QTLs are a major determinant of human expression variation. Nature 482, 390–394 (2012).

    Article  CAS  Google Scholar 

  62. Yang, J., Lee, S. H., Goddard, M. E. & Visscher, P. M. GCTA: a tool for genome-wide complex trait analysis. Am. J. Hum. Genet. 88, 76–82 (2011).

    Article  CAS  Google Scholar 

  63. Storey, J. D. & Tibshirani, R. Statistical significance for genomewide studies. Proc. Natl Acad. Sci. USA 100, 9440–9445 (2003).

    Article  CAS  Google Scholar 

  64. Nica, A. C. et al. The architecture of gene regulatory variation across multiple human tissues: the MuTHER study. PLoS Genet. 7, e1002003 (2011).

    Article  CAS  Google Scholar 

  65. Giulietti, M. et al. SpliceAid-F: a database of human splicing factors and their RNA-binding sites. Nucleic Acids Res. 41, D125–D131 (2013).

    Article  CAS  Google Scholar 

  66. Schmidt, E. M. et al. GREGOR: evaluating global enrichment of trait-associated variants in epigenomic features using a systematic, data-driven approach. Bioinformatics 31, 2601–2606 (2015).

    Article  CAS  Google Scholar 

  67. Lage, K. et al. A human phenome–interactome network of protein complexes implicated in genetic disorders. Nat. Biotechnol. 25, 309–316 (2007).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank the participants of ROS and MAP for their essential contributions and gift to these projects; A. Gusev for helpful discussions and for sharing the source code and scripts for TWAS; the International Genomics of Alzheimer’s Project (IGAP) for providing summary results data for these analyses. This work was supported in part through the computational resources and staff expertise provided by Scientific Computing at the Icahn School of Medicine at Mount Sinai. T.R. is supported by grants from the NIH National Institute on Aging (R01AG054005) and the Alzheimer’s Association. P.L.D. is supported by NIH R01AG036836. D.A.B. is supported by NIH P30AG10161, R01AG015819, R01AG017917. P.L.D. and D.A.B. are supported by NIH U01AG046152. B.Z. is supported by NIH R01AG046170, RF1AG054014, RF1AG057440 and R01AG057907. We thank the patients and families who donated material for CommonMind Consortium data. The CommonMind Consortium data are available in CMC Knowledge Portal: https://www.synapse.org/#!Synapse:syn4923029. Data were generated as part of the CMC supported by funding from Takeda Pharmaceuticals Company Limited, F. Hoffman-La Roche Ltd and NIH grants R01MH085542, R01MH093725, P50MH066392, P50MH080405, R01MH097276, RO1-MH-075916, P50M096891, P50MH084053S1, R37MH057881 and R37MH057881S1, HHSN271201300031C, AG02219, AG05138 and MH06692. Brain tissue for the study was obtained from the following brain bank collections: the Mount Sinai NIH Brain and Tissue Repository, the University of Pennsylvania Alzheimer’s Disease Core Center, the University of Pittsburgh NeuroBioBank and Brain and Tissue Repositories and the NIMH Human Brain Collection Core. CMC Leadership: P.S., J. Buxbaum (Icahn School of Medicine at Mount Sinai), B. Devlin, D. Lewis (University of Pittsburgh), R. Gur, C.-G. Hahn (University of Pennsylvania), K. Hirai, H. Toyoshiba (Takeda Pharmaceuticals Company Limited), E. Domenici, L. Essioux (F. Hoffman-La Roche Ltd), L. Mangravite, M. Peters (Sage Bionetworks), T. Lehner and B. Lipska (NIMH).

Author information

Authors and Affiliations

Authors

Contributions

T.R. and P.L.D. conceived the project and planned the experiments. T.R. and Y.I.L. analyzed and interpreted the data with support from G.W., S.R., J.H., Y.W., I.G., B.N. and S.M. P.L.D., D.A.B., M.W., P.S., E.E.S., V.H. and B.Z. contributed samples and/or data. T.Y.P. performed the tau overexpression in iPSC neurons. T.R. and P.L.D. prepared the first draft of the manuscript. All authors contributed to the final manuscript.

Corresponding authors

Correspondence to Towfique Raj or Philip L. De Jager.

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.

Supplementary Information

Supplementary Text and Figures

Supplementary Figures 1–17 and Supplementary Note

Reporting Summary

Supplementary Table 1

Demographic characteristics of the ROS and MAP cohort

Supplementary Table 2

List of significantly differentially spliced introns associated with neuropathologies

Supplementary Table 3

List of significantly differentially spliced introns associated with clinical AD status

Supplementary Tables 4–7

Supplementary Tables 4–7

Supplementary Table 8

List of differentially spliced introns associated with clinical AD status in ROS/MAP that replicate in the MSBB dataset

Supplementary Table 9

List of differentially spliced introns from overexpressing Tau in iPSC-derived neurons

Supplementary Table 10

List of splicing QTLs at FDR 0.05 identified in the ROS/MAP dataset

Supplementary Table 11

Significant TWAS genes with association to IGAP AD GWAS

Supplementary Table 12

Significant TWAS genes with association to meta-analysis of IGAP and UKBB AD GWAS

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Raj, T., Li, Y.I., Wong, G. et al. Integrative transcriptome analyses of the aging brain implicate altered splicing in Alzheimer’s disease susceptibility. Nat Genet 50, 1584–1592 (2018). https://doi.org/10.1038/s41588-018-0238-1

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41588-018-0238-1

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