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.

  • Technical Report
  • Published:

A statistical framework for cross-tissue transcriptome-wide association analysis

Abstract

Transcriptome-wide association analysis is a powerful approach to studying the genetic architecture of complex traits. A key component of this approach is to build a model to impute gene expression levels from genotypes by using samples with matched genotypes and gene expression data in a given tissue. However, it is challenging to develop robust and accurate imputation models with a limited sample size for any single tissue. Here, we first introduce a multi-task learning method to jointly impute gene expression in 44 human tissues. Compared with single-tissue methods, our approach achieved an average of 39% improvement in imputation accuracy and generated effective imputation models for an average of 120% more genes. We describe a summary-statistic-based testing framework that combines multiple single-tissue associations into a powerful metric to quantify the overall gene–trait association. We applied our method, called UTMOST (unified test for molecular signatures), to multiple genome-wide-association results and demonstrate its advantages over single-tissue strategies.

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: UTMOST workflow.
Fig. 2: Improvement in gene expression imputation accuracy.
Fig. 3: Cross-tissue analysis improves statistical power.
Fig. 4: UTMOST identified more associations in biologically relevant tissues for 50 complex traits.
Fig. 5: Multi-tissue analysis identifies more associations for LDL-C.
Fig. 6: Manhattan plot for LOAD meta-analysis.

Similar content being viewed by others

Data availability

All data used in the manuscript are publicly available (see URLs). GTEx and GERA data can be accessed by application to dbGaP. CommonMind data are available through formal application to NIMH. ADGC phase 2 summary statistics used for validation are available through the NIAGADS portal under accession number NG00076.

References

  1. Visscher, P. M. et al. 10 years of GWAS discovery: biology, function, and translation. Am. J. Hum. Genet. 101, 5–22 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Boyle, E. A., Li, Y. I. & Pritchard, J. K. An Expanded view of complex traits: from polygenic to omnigenic. Cell 169, 1177–1186 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  3. Ardlie, K. G. et al. The Genotype-Tissue Expression (GTEx) pilot analysis: multitissue gene regulation in humans. Science 348, 648–660 (2015).

    Google Scholar 

  4. Aguet, F. et al. Genetic effects on gene expression across human tissues. Nature 550, 204–213 (2017).

    Google Scholar 

  5. Yang, F. et al. Identifying cis-mediators for trans-eQTLs across many human tissues using genomic mediation analysis. Genome Res. 27, 1859–1871 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  6. Saha, A. et al. Co-expression networks reveal the tissue-specific regulation of transcription and splicing. Genome Res. 27, 1843–1858 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  7. Mohammadi, P., Castel, S. E., Brown, A. A. & Lappalainen, T. Quantifying the regulatory effect size of cis-acting genetic variation using allelic fold change. Genome Res. 27, 1872–1884 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  8. 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).

    PubMed  PubMed Central  Google Scholar 

  9. Hou, L., Chen, M., Zhang, C. K., Cho, J. & Zhao, H. Guilt by rewiring: gene prioritization through network rewiring in genome wide association studies. Hum. Mol. Genet. 23, 2780–2790 (2013).

    PubMed  PubMed Central  Google Scholar 

  10. Giambartolomei, C. et al. Bayesian test for colocalisation between pairs of genetic association studies using summary statistics. PLoS Genet. 10, e1004383 (2014).

    PubMed  PubMed Central  Google Scholar 

  11. Zhu, Z. et al. Integration of summary data from GWAS and eQTL studies predicts complex trait gene targets. Nat. Genet. 48, 481–487 (2016).

    CAS  PubMed  Google Scholar 

  12. Hormozdiari, F. et al. Colocalization of GWAS and eQTL signals detects target genes. Am. J. Hum. Genet. 99, 1245–1260 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  13. Zhao, S. D., Cai, T. T., Cappola, T. P., Margulies, K. B. & Li, H. Sparse simultaneous signal detection for identifying genetically controlled disease genes. J. Am. Stat. Assoc. 112, 1032–1046 (2016).

    Google Scholar 

  14. Gamazon, E. R. et al. A gene-based association method for mapping traits using reference transcriptome data. Nat. Genet. 47, 1091–1098 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  16. Mancuso, N. et al. Integrating gene expression with summary association statistics to identify genes associated with 30 complex traits. Am. J. Hum. Genet. 100, 473–487 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Barbeira, A. N. et al. Exploring the phenotypic consequences of tissue specific gene expression variation inferred from GWAS summary statistics. Nat. Commun. 9, 1825 (2018).

    PubMed  PubMed Central  Google Scholar 

  18. Hoffman, J. D. et al. Cis-eQTL-based trans-ethnic meta-analysis reveals novel genes associated with breast cancer risk. PLoS Genet. 13, e1006690 (2017).

    PubMed  PubMed Central  Google Scholar 

  19. Liu, X. et al. Functional architectures of local and distal regulation of gene expression in multiple human tissues. Am. J. Hum. Genet. 100, 605–616 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  20. Wainberg, M. et al. Vulnerabilities of transcriptome-wide association studies. Preprint at https://www.biorxiv.org/content/10.1101/206961v5 (2017).

  21. Li, C., Yang, C., Gelernter, J. & Zhao, H. Improving genetic risk prediction by leveraging pleiotropy. Hum. Genet. 133, 639–650 (2014).

    PubMed  Google Scholar 

  22. Maier, R. et al. Joint analysis of psychiatric disorders increases accuracy of risk prediction for schizophrenia, bipolar disorder, and major depressive disorder. Am. J. Hum. Genet. 96, 283–294 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  23. Hu, Y. et al. Joint modeling of genetically correlated diseases and functional annotations increases accuracy of polygenic risk prediction. PLoS Genet. 13, e1006836 (2017).

    PubMed  PubMed Central  Google Scholar 

  24. Flutre, T., Wen, X., Pritchard, J. & Stephens, M. A statistical framework for joint eQTL analysis in multiple tissues. PLoS Genet. 9, e1003486 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  25. Sul, J. H., Han, B., Ye, C., Choi, T. & Eskin, E. Effectively identifying eQTLs from multiple tissues by combining mixed model and meta-analytic approaches. PLoS Genet. 9, e1003491 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  26. Duong, D. et al. Applying meta-analysis to genotype-tissue expression data from multiple tissues to identify eQTLs and increase the number of eGenes. Bioinformatics 33, i67–i74 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  27. Li, G., Jima, D. D., Wright, F. A. & Nobel, A. B. HT-eQTL: integrative eQTL analysis in a large number of human tissues. BMC Bioinformatics 19, 95 (2018).

    PubMed  PubMed Central  Google Scholar 

  28. Hore, V. et al. Tensor decomposition for multiple-tissue gene expression experiments. Nat. Genet. 48, 1094–1100 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  29. Yuan, M. & Lin, Y. Model selection and estimation in regression with grouped variables. J. Royal Stat. Soc. B 68, 49–67 (2006).

    Google Scholar 

  30. Sun, R. & Lin, X. Set-based tests for genetic association using the generalized Berk–Jones statistic. Preprint at https://arxiv.org/pdf/1710.02469 (2017).

  31. Zhou, X., Carbonetto, P. & Stephens, M. Polygenic modeling with Bayesian sparse linear mixed models. PLoS Genet. 9, e1003264 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  32. Lappalainen, T. et al. Transcriptome and genome sequencing uncovers functional variation in humans. Nature 501, 506 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  34. Finucane, H. K. et al. Partitioning heritability by functional annotation using genome-wide association summary statistics. Nat. Genet. 47, 1228 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  35. Lu, Q. et al. Systematic tissue-specific functional annotation of the human genome highlights immune-related DNA elements for late-onset Alzheimer’s disease. PLoS Genet. 13, e1006933 (2017).

    PubMed  PubMed Central  Google Scholar 

  36. Global Lipids Genetics Consortium. Discovery and refinement of loci associated with lipid levels. Nat. Genet. 45, 1274–1283 (2013).

    Google Scholar 

  37. Franzén, O. et al. Cardiometabolic risk loci share downstream cis-and trans-gene regulation across tissues and diseases. Science 353, 827–830 (2016).

    PubMed  PubMed Central  Google Scholar 

  38. Musunuru, K. et al. From noncoding variant to phenotype via SORT1 at the 1p13 cholesterol locus. Nature 466, 714–719 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  39. Strong, A. et al. Hepatic sortilin regulates both apolipoprotein B secretion and LDL catabolism. J. Clin. Invest. 122, 2807 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  40. 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).

    CAS  PubMed  PubMed Central  Google Scholar 

  41. Gagliano, S. A. et al. Genomics implicates adaptive and innate immunity in Alzheimer’s and Parkinson’s diseases. Ann. Clin. Transl. Neurol. 3, 924–933 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  42. Huang, K. L. et al. A common haplotype lowers PU. 1 expression in myeloid cells and delays onset of Alzheimer’s disease. Nat. Neurosci. 20, 1052 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  43. Raj, T. et al. Integrative transcriptome analyses of the aging brain implicate altered splicing in Alzheimer’s disease susceptibility. Nat. Genet. 50, 1584 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  44. Chen, L. et al. Genetic drivers of epigenetic and transcriptional variation in human immune cells. Cell 167, 1398–1414. e24 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  45. 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).

    CAS  PubMed  Google Scholar 

  46. Hollingworth, P. et al. Common variants at ABCA7, MS4A6A/MS4A4E, EPHA1, CD33 and CD2AP are associated with Alzheimer’s disease. Nat. Genet. 43, 429–435 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  47. Harold, D. et al. Genome-wide association study identifies variants at CLU and PICALM associated with Alzheimer’s disease. Nat. Genet. 41, 1088–1093 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  48. Naj, A. C. et al. Common variants at MS4A4/MS4A6E, CD2AP, CD33 and EPHA1 are associated with late-onset Alzheimer’s disease. Nat. Genet. 43, 436–441 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  49. Seshadri, S. et al. Genome-wide analysis of genetic loci associated with Alzheimer disease. JAMA 303, 1832–1840 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  50. Jun, G. R. et al. Transethnic genome-wide scan identifies novel Alzheimer’s disease loci. Alzheimers Dement. 13, 727–738 (2017).

    PubMed  PubMed Central  Google Scholar 

  51. Lambert, J. C. et al. Genome-wide association study identifies variants at CLU and CR1 associated with Alzheimer’s disease. Nat. Genet. 41, 1094–1099 (2009).

    CAS  PubMed  Google Scholar 

  52. Sherva, R. et al. Genome-wide association study of the rate of cognitive decline in Alzheimer’s disease. Alzheimers Dement. 10, 45–52 (2014).

    PubMed  Google Scholar 

  53. Crehan, H. et al. Complement receptor 1 (CR1) and Alzheimer’s disease. Immunobiology 217, 244–250 (2012).

    CAS  PubMed  Google Scholar 

  54. Liu, J. Z. et al. Association analyses identify 38 susceptibility loci for inflammatory bowel disease and highlight shared genetic risk across populations. Nat. Genet. 47, 979–986 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  55. Remmers, E. F. et al. Genome-wide association study identifies variants in the MHC class I, IL10, and IL23R-IL12RB2 regions associated with Behcet’s disease. Nat. Genet. 42, 698–702 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  56. Plagnol, V. et al. Genome-wide association analysis of autoantibody positivity in type 1 diabetes cases. PLoS Genet. 7, e1002216 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  57. Bentham, J. et al. Genetic association analyses implicate aberrant regulation of innate and adaptive immunity genes in the pathogenesis of systemic lupus erythematosus. Nat. Genet. 47, 1457–1464 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  58. Kiyota, T. et al. AAV serotype 2/1-mediated gene delivery of anti-inflammatory interleukin-10 enhances neurogenesis and cognitive function in APP + PS1 mice. Gene Ther. 19, 724–733 (2012).

    CAS  PubMed  Google Scholar 

  59. Chakrabarty, P. et al. IL-10 alters immunoproteostasis in APP mice, increasing plaque burden and worsening cognitive behavior. Neuron 85, 519–533 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  60. Xu, M. et al. A systematic integrated analysis of brain expression profiles reveals YAP1 and other prioritized hub genes as important upstream regulators in Alzheimer’s disease. Alzheimers Dement. 14, 215–229 (2017).

    CAS  PubMed  Google Scholar 

  61. Hohman, T. J. et al. Discovery of gene–gene interactions across multiple independent data sets of late onset Alzheimer disease from the Alzheimer Disease Genetics Consortium. Neurobiol. Aging 38, 141–150 (2016).

    CAS  PubMed  Google Scholar 

  62. Katsouri, L. et al. Prazosin, an α 1-adrenoceptor antagonist, prevents memory deterioration in the APP23 transgenic mouse model of Alzheimer’s disease. Neurobiol. Aging 34, 1105–1115 (2013).

    CAS  PubMed  Google Scholar 

  63. Duplan, L. et al. Lithostathine and pancreatitis-associated protein are involved in the very early stages of Alzheimer’s disease. Neurobiol. Aging 22, 79–88 (2001).

    CAS  PubMed  Google Scholar 

  64. Stenmark, H. & Olkkonen, V. M. The rab gtpase family. Genome. Biol. 2, reviews3007 (2001).

    CAS  PubMed  PubMed Central  Google Scholar 

  65. Lin, B. D. et al. Heritability and GWAS studies for monocyte–lymphocyte ratio. Twin Res Hum. Genet. 20, 97–107 (2017).

    PubMed  Google Scholar 

  66. Astle, W. J. et al. The allelic landscape of human blood cell trait variation and links to common complex disease. Cell 167, 1415–1429.e19 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  67. Li, T. et al. Identification of the gene for vitamin K epoxide reductase. Nature 427, 541–544 (2004).

    CAS  PubMed  Google Scholar 

  68. Kohnke, H., Sörlin, K., Granath, G. & Wadelius, M. Warfarin dose related to apolipoprotein E (APOE) genotype. Eur. J. Clin. Pharmacol. 61, 381–388 (2005).

    CAS  PubMed  Google Scholar 

  69. Teslovich, T. M. et al. Biological, clinical and population relevance of 95 loci for blood lipids. Nature 466, 707–713 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  70. de Lange, K. M. et al. Genome-wide association study implicates immune activation of multiple integrin genes in inflammatory bowel disease. Nat. Genet. 49, 256–261 (2017).

    PubMed  PubMed Central  Google Scholar 

  71. Davies, G. et al. Genetic contributions to variation in general cognitive function: a meta-analysis of genome-wide association studies in the CHARGE consortium (N = 53 949). Mol. Psychiatry 20, 183 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  72. Torres, J. M. et al. Integrative cross tissue analysis of gene expression identifies novel type 2 diabetes genes. Preprint at https://www.biorxiv.org/content/10.1101/108134v2 (2017).

  73. Park, Y. et al. Causal gene inference by multivariate mediation analysis in Alzheimer’s disease. Preprint at https://www.biorxiv.org/content/10.1101/219428v3 (2017).

  74. Mancuso, N. et al. Probabilistic fine-mapping of transcriptome-wide association studies. Preprint at https://www.biorxiv.org/content/10.1101/236869v2 (2017).

  75. Xu, Z., Wu, C., Wei, P. & Pan, W. A powerful framework for integrating eQTL and GWAS summary data. Genetics 207, 893–902 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  76. GTEx Consortium et al. Genetic effects on gene expression across human tissues. Nature 550, 204–213 (2017).

  77. Stegle, O., Parts, L., Durbin, R. & Winn, J. A Bayesian framework to account for complex non-genetic factors in gene expression levels greatly increases power in eQTL studies. PLoS Comput. Biol. 6, e1000770 (2010).

    PubMed  PubMed Central  Google Scholar 

  78. O’Connor, L. J. et al. Estimating the proportion of disease heritability mediated by gene expression levels. Preprint at https://www.biorxiv.org/content/10.1101/118018v1 (2017).

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

    CAS  PubMed  PubMed Central  Google Scholar 

  80. Marchini, J., Howie, B., Myers, S., McVean, G. & Donnelly, P. A new multipoint method for genome-wide association studies by imputation of genotypes. Nat. Genet. 39, 906–913 (2007).

    CAS  PubMed  Google Scholar 

  81. Willer, C. J., Li, Y. & Abecasis, G. R. METAL: fast and efficient meta-analysis of genomewide association scans. Bioinformatics 26, 2190–2191 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  82. Kundaje, A. et al. Integrative analysis of 111 reference human epigenomes. Nature 518, 317 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  83. Lu, Q. et al. Systematic tissue-specific functional annotation of the human genome highlights immune-related DNA elements for late-onset Alzheimer’s disease. PLoS Genet. 13, e1006933 (2017).

    PubMed  PubMed Central  Google Scholar 

  84. Finucane, H. K. et al. Heritability enrichment of specifically expressed genes identifies disease-relevant tissues and cell types. Nat. Genet. 50, 621 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  85. Abecasis, G. R. et al. An integrated map of genetic variation from 1,092 human genomes. Nature 491, 56–65 (2012).

    PubMed  Google Scholar 

  86. Huang, D. W., Sherman, B. T. & Lempicki, R. A. Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources. Nat. Protoc. 4, 44–57 (2009).

    CAS  Google Scholar 

  87. Pruim, R. J. et al. LocusZoom: regional visualization of genome-wide association scan results. Bioinformatics 26, 2336–2337 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  88. Turner, S. D. qqman: an R package for visualizing GWAS results using QQ and manhattan plots. Preprint at https://www.biorxiv.org/content/10.1101/005165v1 (2014).

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

    CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

This study was supported in part by NIH grants R01 GM59507 and 3P30AG021342-16S2 (Y.H., M.L., Q.L., and H.Z.), CTSA UL1TR000427 (Q.L.), R01 AG042437 and U01 AG006781 (P.K.C. and S. Mukherjee); the Yale World Scholars Program sponsored by the China Scholarship Council (J.W., and Z.Y.); Neil Shen’s SJTU Medical Research Fund, the SJTU-Yale Collaborative Research Seed Fund; and NSFC 31728012 (J.G., H.L., and H.Z.), and the National Key R&D Program of China 2018YFC0910500 (J.G., and H.L). We thank C. Brown for assistance in matching GTEx tissues to Roadmap cell types. This study makes use of summary statistics from many GWAS consortia. We thank the investigators in these GWAS consortia for generously sharing their data. We thank the IGAP for providing summary results data for these analyses. The investigators within IGAP contributed to the design and implementation of IGAP and/or provided data but did not participate in the analysis or writing of this report. IGAP was made possible by the generous participation of the subjects and their families. The i-Select chips were funded by the French National Foundation on Alzheimer’s disease and related disorders. EADI was supported by the LABEX (Laboratory of Excellence Program Investment for the Future) DISTALZ grant, Inserm, Institut Pasteur de Lille, Université de Lille 2, and the Lille University Hospital. The Genetic and Environmental Risk in AD consortium (GERAD) was supported by the Medical Research Council (grant no. 503480), Alzheimer’s Research UK (grant no. 503176), the Wellcome Trust (grant no. 082604/2/07/Z), and the German Federal Ministry of Education and Research (BMBF): Competence Network Dementia (CND) grant nos. 01GI0102, 01GI0711, and 01GI0420. The Cohorts for Heart and Aging Research in Genomic Epidemiology consortium (CHARGE) was partly supported by NIH/NIA grant no. R01 AG033193, NIA grant no. AG081220, AGES contract N01–AG–12100, NHLBI grant no. R01 HL105756, the Icelandic Heart Association, and the Erasmus Medical Center and Erasmus University. ADGC was supported by NIH/NIA grants nos. U01 AG032984, U24 AG021886, and U01 AG016976, and the Alzheimer’s Association grant no. ADGC–10–196728. We thank the contributors who collected the samples used in this study, as well as the patients and their families, whose help and participation made this work possible; data for this study were prepared, archived, and distributed by the National Institute on Aging Alzheimer’s Disease Data Storage Site (NIAGADS) at the University of Pennsylvania (U24-AG041689-01). We are also grateful for all the consortia and investigators that provided publicly accessible GWAS summary statistics.

Author information

Authors and Affiliations

Authors

Consortia

Contributions

Y.H., M.L., Q.L., H.L., and H.Z. conceived the study and developed the statistical model. Y.H., M.L., Q.L., H.W., J.W., S.M.Z., B.L., Y.S., S. Muchnik, and J.G. performed the statistical analyses. S.M.Z. and P.N. assisted in LDL analysis. Y.H., M.L., Z.Y., and Q.L. implemented the software. B.W.K. prepared ADGC summary statistics. A.N., A.K., and Y.Z. assisted in data preparation. S. Mukherjee and P.K.C. assisted in Alzheimer’s disease data application, curation, and interpretation. Y.H., M.L., Q.L., H.L., and H.Z. wrote the manuscript. H.Z. advised on statistical and genetic issues. All authors contributed to manuscript editing and approved the manuscript.

Corresponding author

Correspondence to Hongyu Zhao.

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 Note, Supplementary Tables 1–13, 21–23 and 26–29, and Supplementary Figures 1–13

Reporting Summary

Supplementary Tables

Supplementary Tables 14–20, 24 and 25

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Hu, Y., Li, M., Lu, Q. et al. A statistical framework for cross-tissue transcriptome-wide association analysis. Nat Genet 51, 568–576 (2019). https://doi.org/10.1038/s41588-019-0345-7

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41588-019-0345-7

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