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.

  • Article
  • Published:

Modeling tissue co-regulation estimates tissue-specific contributions to disease

Abstract

Integrative analyses of genome-wide association studies and gene expression data have implicated many disease-critical tissues. However, co-regulation of genetic effects on gene expression across tissues impedes distinguishing biologically causal tissues from tagging tissues. In the present study, we introduce tissue co-regulation score regression (TCSC), which disentangles causal tissues from tagging tissues by regressing gene–disease association statistics (from transcriptome-wide association studies) on tissue co-regulation scores, reflecting correlations of predicted gene expression across genes and tissues. We applied TCSC to 78 diseases/traits (average n = 302,000) and gene expression prediction models for 48 GTEx tissues. TCSC identified 21 causal tissue–trait pairs at a 5% false discovery rate (FDR), including well-established findings, biologically plausible new findings (for example, aorta artery and glaucoma) and increased specificity of known tissue–trait associations (for example, subcutaneous adipose, but not visceral adipose, and high-density lipoprotein). TCSC also identified 17 causal tissue–trait covariance pairs at 5% FDR. In conclusion, TCSC is a precise method for distinguishing causal tissues from tagging tissues.

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: Overview of TCSC regression.
Fig. 2: Robustness and power of TCSC regression in simulations.
Fig. 3: TCSC estimates tissue-specific contributions to disease and complex trait heritability.
Fig. 4: Comparison of disease-critical tissues identified by RTC Coloc, LDSC-SEG and TCSC.
Fig. 5: Cross-trait TCSC estimates tissue-specific contributions to the genetic covariance of two diseases/traits.

Similar content being viewed by others

Data availability

We have made 78 GWAS summary statistics and 41 brain-specific summary statistics publicly available at https://github.com/TiffanyAmariuta/TCSC/tree/main/sumstats (https://doi.org/10.5281/zenodo.8030594)73, gene expression prediction models publicly available at https://alkesgroup.broadinstitute.org/TCSC/GeneExpressionModels, TWAS association statistics publicly available at https://alkesgroup.broadinstitute.org/TCSC/TWAS_sumstats, tissue co-regulation scores publicly available at https://github.com/TiffanyAmariuta/TCSC/tree/main/coregulation_scores and TCSC output publicly available at https://github.com/TiffanyAmariuta/TCSC/tree/main/results. Gene expression and genotype data were acquired from the GTEx v.8 eQTL dataset (dbGaP, accession no. phs000424.v8.p2) and 1000 Genomes phase 3 data were downloaded from https://data.broadinstitute.org/alkesgroup/FUSION/LDREF.tar.bz2. Source data are provided with this paper.

Code availability

TCSC software including a quick start tutorial is available at https://github.com/TiffanyAmariuta/TCSC/ (https://doi.org/10.5281/zenodo.8030594)73. The Mancuso Lab TWAS Simulator is available at https://github.com/mancusolab/twas_sim. The FUSION software is available at http://gusevlab.org/projects/fusion. The simulation code for RTC Coloc is available at https://github.com/TiffanyAmariuta/TCSC/tree/main/simulation_analysis.

References

  1. Hekselman, I. & Yeger-Lotem, E. Mechanisms of tissue and cell-type specificity in heritable traits and diseases. Nat. Rev. Genet. 21, 137–150 (2020).

    Article  CAS  PubMed  Google Scholar 

  2. Ongen, H. et al. Estimating the causal tissues for complex traits and diseases. Nat. Genet. 49, 1676–1683 (2017).

    Article  CAS  PubMed  Google Scholar 

  3. Gamazon, E. R. et al. Using an atlas of gene regulation across 44 human tissues to inform complex disease- and trait-associated variation. Nat. Genet. 50, 956–967 (2018).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  4. Hormozdiari, F. et al. Leveraging molecular quantitative trait loci to understand the genetic architecture of diseases and complex traits. Nat. Genet. 50, 1041–1047 (2018).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  5. Arvanitis, M., Tayeb, K., Strober, B. J. & Battle, A. Redefining tissue specificity of genetic regulation of gene expression in the presence of allelic heterogeneity. Am. J. Hum. Genet. 109, 223–239 (2022).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  6. Calderon, D. et al. Inferring relevant cell types for complex traits by using single-cell gene expression. Am. J. Hum. Genet. 101, 686–699 (2017).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  7. Finucane, H. K. et al. Heritability enrichment of specifically expressed genes identifies disease-relevant tissues and cell types. Nat. Genet. 50, 621–629 (2018).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  8. Bryois, J. et al. Genetic identification of cell types underlying brain complex traits yields insights into the etiology of Parkinson’s disease. Nat. Genet. 52, 482–493 (2020).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  9. Shang, L., Smith, J. A. & Zhou, X. Leveraging gene co-expression patterns to infer trait-relevant tissues in genome-wide association studies. PLoS Genet. 16, e1008734 (2020).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  10. Maurano, M. T. et al. Systematic localization of common disease-associated variation in regulatory DNA. Science 337, 1190–1195 (2012).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  11. Trynka, G. et al. Chromatin marks identify critical cell types for fine mapping complex trait variants. Nat. Genet. 45, 124–130 (2013).

    Article  CAS  PubMed  Google Scholar 

  12. Pickrell, J. K. Joint analysis of functional genomic data and genome-wide association studies of 18 human traits. Am. J. Hum. Genet. 94, 559–573 (2014).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  13. Finucane, H. K. et al. Partitioning heritability by functional annotation using genome-wide association summary statistics. Nat. Genet. 47, 1228–1235 (2015).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  14. Roadmap Epigenomics Consortium et al. Integrative analysis of 111 reference human epigenomes. Nature 518, 317–330 (2015).

    Article  PubMed Central  Google Scholar 

  15. Backenroth, D. et al. FUN-LDA: a latent dirichlet allocation model for predicting tissue-specific functional effects of noncoding variation: methods and applications. Am. J. Hum. Genet. 102, 920–942 (2018).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  16. Amariuta, T. et al. IMPACT: genomic annotation of cell-state-specific regulatory elements inferred from the epigenome of bound transcription factors. Am. J. Hum. Genet. 104, 879–895 (2019).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  17. Boix, C. A., James, B. T., Park, Y. P., Meuleman, W. & Kellis, M. Regulatory genomic circuitry of human disease loci by integrative epigenomics. Nature 590, 300–307 (2021).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  18. Wainberg, M. et al. Opportunities and challenges for transcriptome-wide association studies. Nat. Genet. 51, 592–599 (2019).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  19. The GTEx Consortium. The GTEx Consortium atlas of genetic regulatory effects across human tissues. Science 369, 1318–1330 (2020).

    Article  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed Central  PubMed  Google Scholar 

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

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  22. Siewert-Rocks, K. M., Kim, S. S., Yao, D. W., Shi, H. & Price, A. L. Leveraging gene co-regulation to identify gene sets enriched for disease heritability. Am. J. Hum. Genet. 109, 393–404 (2022).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  23. Schaid, D. J., Chen, W. & Larson, N. B. From genome-wide associations to candidate causal variants by statistical fine-mapping. Nat. Rev. Genet. 19, 491–504 (2018).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  24. Zhu, H., Shang, L. & Zhou, X. A review of statistical methods for identifying trait-relevant tissues and cell types. Front. Genet. 11, 587887 (2020).

    Article  PubMed  Google Scholar 

  25. Bulik-Sullivan, B. K. et al. LD score regression distinguishes confounding from polygenicity in genome-wide association studies. Nat. Genet. 47, 291–295 (2015).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  26. Mancuso, N. et al. Probabilistic fine-mapping of transcriptome-wide association studies. Nat. Genet. 51, 675–682 (2019).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  27. The 1000 Genomes Project Consortium. An integrated map of genetic variation from 1,092 human genomes. Nature 491, 56–65 (2012).

    Article  PubMed Central  Google Scholar 

  28. Gazal, S. et al. Linkage disequilibrium-dependent architecture of human complex traits shows action of negative selection. Nat. Genet. 49, 1421–1427 (2017).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  29. Gazal, S., Marquez-Luna, C., Finucane, H. K. & Price, A. L. Reconciling S-LDSC and LDAK functional enrichment estimates. Nat. Genet. 51, 1202–1204 (2019).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  30. Homan, T. D., Bordes, S. & Cichowski, E. Physiology, pulse pressure. in StatPearls (Treasure Island, 2022).

  31. Kass, M. A. et al. The ocular hypertension treatment study: a randomized trial determines that topical ocular hypotensive medication delays or prevents the onset of primary open-angle glaucoma. Arch. Ophthalmol. 120, 701–713 (2002).

    Article  PubMed  Google Scholar 

  32. Zhao, D., Cho, J., Kim, M. H. & Guallar, E. The association of blood pressure and primary open-angle glaucoma: a meta-analysis. Am. J. Ophthalmol. 158, 615–27 e9 (2014).

    Article  PubMed  Google Scholar 

  33. Levine, R. M., Yang, A., Brahma, V. & Martone, J. F. Management of blood pressure in patients with glaucoma. Curr. Cardiol. Rep. 19, 109 (2017).

    Article  PubMed  Google Scholar 

  34. De Moraes, C. G., Cioffi, G. A., Weinreb, R. N. & Liebmann, J. M. New recommendations for the treatment of systemic hypertension and their potential implications for glaucoma management. J. Glaucoma 27, 567–571 (2018).

    Article  PubMed Central  PubMed  Google Scholar 

  35. Leeman, M. & Kestelyn, P. Glaucoma and blood pressure. Hypertension 73, 944–950 (2019).

    Article  CAS  PubMed  Google Scholar 

  36. Gregg, D. & Goldschmidt-Clermont, P. J. Cardiology patient page. Platelets and cardiovascular disease. Circulation 108, e88–e90 (2003).

    Article  CAS  PubMed  Google Scholar 

  37. Coppinger, J. A. et al. Characterization of the proteins released from activated platelets leads to localization of novel platelet proteins in human atherosclerotic lesions. Blood 103, 2096–2104 (2004).

    Article  CAS  PubMed  Google Scholar 

  38. Gawaz, M., Langer, H. & May, A. E. Platelets in inflammation and atherogenesis. J. Clin. Invest. 115, 3378–3384 (2005).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  39. Davi, G. & Patrono, C. Platelet activation and atherothrombosis. N. Engl. J. Med. 357, 2482–2494 (2007).

    Article  CAS  PubMed  Google Scholar 

  40. Meadows, T. A. & Bhatt, D. L. Clinical aspects of platelet inhibitors and thrombus formation. Circ. Res. 100, 1261–1275 (2007).

    Article  CAS  PubMed  Google Scholar 

  41. Berman, M. N., Tupper, C. & Bhardwaj, A. Physiology, left ventricular function. in StatPearls (Treasure Island, 2022).

  42. Chung, S., Sawyer, J. K., Gebre, A. K., Maeda, N. & Parks, J. S. Adipose tissue ATP binding cassette transporter A1 contributes to high-density lipoprotein biogenesis in vivo. Circulation 124, 1663–1672 (2011).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  43. McGillicuddy, F. C., Reilly, M. P. & Rader, D. J. Adipose modulation of high-density lipoprotein cholesterol: implications for obesity, high-density lipoprotein metabolism, and cardiovascular disease. Circulation 124, 1602–1605 (2011).

    Article  PubMed Central  PubMed  Google Scholar 

  44. Zoccali, C. et al. Adiponectin, metabolic risk factors, and cardiovascular events among patients with end-stage renal disease. J. Am. Soc. Nephrol. 13, 134–141 (2002).

    Article  CAS  PubMed  Google Scholar 

  45. Ryo, M. et al. Adiponectin as a biomarker of the metabolic syndrome. Circ. J. 68, 975–981 (2004).

    Article  CAS  PubMed  Google Scholar 

  46. Toth, P. P. Adiponectin and high-density lipoprotein: a metabolic association through thick and thin. Eur. Heart J. 26, 1579–1581 (2005).

    Article  CAS  PubMed  Google Scholar 

  47. Van Linthout, S. et al. Impact of HDL on adipose tissue metabolism and adiponectin expression. Atherosclerosis 210, 438–444 (2010).

    Article  PubMed  Google Scholar 

  48. Shungin, D. et al. New genetic loci link adipose and insulin biology to body fat distribution. Nature 518, 187–196 (2015).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  49. Emdin, C. A. et al. Genetic association of waist-to-hip ratio with cardiometabolic traits, type 2 diabetes, and coronary heart disease. J. Am. Med. Assoc. 317, 626–634 (2017).

    Article  Google Scholar 

  50. Smith, J., Al-Amri, M., Sniderman, A. & Cianflone, K. Leptin and adiponectin in relation to body fat percentage, waist to hip ratio and the apoB/apoA1 ratio in Asian Indian and Caucasian men and women. Nutr. Metab. 3, 18 (2006).

    Article  Google Scholar 

  51. Farooqi, I. S. Defining the neural basis of appetite and obesity: from genes to behaviour. Clin. Med. 14, 286–289 (2014).

    Article  Google Scholar 

  52. Locke, A. E. et al. Genetic studies of body mass index yield new insights for obesity biology. Nature 518, 197–206 (2015).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  53. Medic, N. et al. Increased body mass index is associated with specific regional alterations in brain structure. Int. J. Obes. 40, 1177–1182 (2016).

    Article  CAS  Google Scholar 

  54. Yengo, L. et al. Meta-analysis of genome-wide association studies for height and body mass index in approximately 700000 individuals of European ancestry. Hum. Mol. Genet. 27, 3641–3649 (2018).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  55. Onat, F. & Cavdar, S. Cerebellar connections: hypothalamus. Cerebellum 2, 263–269 (2003).

    Article  PubMed  Google Scholar 

  56. Zhu, J. N. & Wang, J. J. The cerebellum in feeding control: possible function and mechanism. Cell Mol. Neurobiol. 28, 469–478 (2008).

    Article  PubMed  Google Scholar 

  57. Low, A. Y. T. et al. Reverse-translational identification of a cerebellar satiation network. Nature 600, 269–273 (2021).

    Article  CAS  PubMed  Google Scholar 

  58. Floresco, S. B., Todd, C. L. & Grace, A. A. Glutamatergic afferents from the hippocampus to the nucleus accumbens regulate activity of ventral tegmental area dopamine neurons. J. Neurosci. 21, 4915–4922 (2001).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  59. Enard, W. et al. A humanized version of Foxp2 affects cortico-basal ganglia circuits in mice. Cell 137, 961–971 (2009).

    Article  CAS  PubMed  Google Scholar 

  60. Demontis, D. et al. Discovery of the first genome-wide significant risk loci for attention deficit/hyperactivity disorder. Nat. Genet. 51, 63–75 (2019).

    Article  CAS  PubMed  Google Scholar 

  61. Bulik-Sullivan, B. et al. An atlas of genetic correlations across human diseases and traits. Nat. Genet. 47, 1236–1241 (2015).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  62. Wei, H. S. et al. Erythrocytes are oxygen-sensing regulators of the cerebral microcirculation. Neuron 91, 851–862 (2016).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  63. Olusi, S. O. Obesity is an independent risk factor for plasma lipid peroxidation and depletion of erythrocyte cytoprotectic enzymes in humans. Int. J. Obes. Relat. Metab. Disord. 26, 1159–1164 (2002).

    Article  CAS  PubMed  Google Scholar 

  64. Ozata, M. et al. Increased oxidative stress and hypozincemia in male obesity. Clin. Biochem. 35, 627–631 (2002).

    Article  CAS  PubMed  Google Scholar 

  65. Druml, W., Laggner, A. N., Lenz, K., Grimm, G. & Schneeweiss, B. Pancreatitis in acute hemolysis. Ann. Hematol. 63, 39–41 (1991).

    Article  CAS  PubMed  Google Scholar 

  66. Sakai, N. S., Taylor, S. A. & Chouhan, M. D. Obesity, metabolic disease and the pancreas—quantitative imaging of pancreatic fat. Br. J. Radiol. 91, 20180267 (2018).

    Article  PubMed Central  PubMed  Google Scholar 

  67. Peterson, C. G., Skoog, V. & Venge, P. Human eosinophil cationic proteins (ECP and EPX) and their suppressive effects on lymphocyte proliferation. Immunobiology 171, 1–13 (1986).

    Article  CAS  PubMed  Google Scholar 

  68. Nakagome, K. et al. IL-5-induced hypereosinophilia suppresses the antigen-induced immune response via a TGF-beta-dependent mechanism. J. Immunol. 179, 284–294 (2007).

    Article  CAS  PubMed  Google Scholar 

  69. Onyema, O. O. et al. Eosinophils downregulate lung alloimmunity by decreasing TCR signal transduction. JCI Insight 4, e128241 (2019).

    Article  PubMed Central  PubMed  Google Scholar 

  70. Yao, D. W., O’Connor, L. J., Price, A. L. & Gusev, A. Quantifying genetic effects on disease mediated by assayed gene expression levels. Nat. Genet. 52, 626–633 (2020).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  71. Weissbrod, O. et al. Leveraging fine-mapping and multipopulation training data to improve cross-population polygenic risk scores. Nat. Genet. 54, 450–458 (2022).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  72. 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).

    Article  PubMed Central  PubMed  Google Scholar 

  73. Amariuta, T. TCSC GitHub Repository (2023); https://doi.org/10.5281/zenodo.8030594

  74. Bycroft, C. et al. The UK Biobank resource with deep phenotyping and genomic data. Nature 562, 203–209 (2018).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  75. Gazal, S. et al. Functional architecture of low-frequency variants highlights strength of negative selection across coding and non-coding annotations. Nat. Genet. 50, 1600–1607 (2018).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  76. 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  PubMed Central  PubMed  Google Scholar 

  77. Reshef, Y. A. et al. Detecting genome-wide directional effects of transcription factor binding on polygenic disease risk. Nat. Genet. 50, 1483–1493 (2018).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  78. Zhang, M. J. et al. Polygenic enrichment distinguishes disease associations of individual cells in single-cell RNA-seq data. Nat. Genet. 54, 1572–1580 (2022).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

Download references

Acknowledgements

We thank H. Shi, M. Zhang and B. Strober for helpful discussions. This work was funded by US National Institutes of Health grants (nos. U01 HG009379, R01 MH101244, R37 MH107649, R01 HG006399, R01 MH115676 and U01 HG012009 awarded to A.L.P.). The funders had no role in study design, data collection and analysis, decision to publish or preparation of the manuscript.

Author information

Authors and Affiliations

Authors

Contributions

T.A. and A.L.P. conceived and designed the study. T.A. conducted simulation and real data analysis. K.S.-W. helped design the simulation framework. T.A. and A.L.P. wrote the initial draft of the manuscript. All authors contributed to the final manuscript.

Corresponding authors

Correspondence to Tiffany Amariuta or Alkes L. Price.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Genetics thanks Xia Shen and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

Additional information

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

Extended data

Extended Data Fig. 1 Comparison of tissue-trait association methods with TCSC in simulations.

(a) Percentage of estimates of \({h}_{ge(t^{\prime} )}^{2}\) for noncausal tissues that were significantly positive at P < 0.05. \({h}_{ge(t\,causal)}^{2}\) is set to 10% and GWAS sample size is set to 10,000. (b) Percentage of estimates of \({h}_{ge(t{\prime} )}^{2}\) for causal tissues that were significantly positive at p < 0.05. In panels A and B, we performed n = 1,000 independent simulated genetic architectures for TCSC, LDSC-SEG, CoCoNet, and RolyPoly and n = 100 independent simulated genetic architectures for RTC Coloc due to the computationally intensive nature of the method across each eQTL sample size (n = 100, 200, 300, 500, 1000, 1500); we used a one-sided z-test and the genomic block jackknife standard error to obtain p-values and data are presented as mean values ± 1.96 × s.e.m. (c) Receiver operating characteristic (ROC) curves for each method, including cross-trait TCSC, across 1,000 uniformly spaced p-values between 0 and 1 used as the threshold to identify a causal tissue at a simulation eQTL sample size of 300, most closely matching real data analysis. We note that CoCoNet cannot be compared here because the method identifies the single most likely causal tissue using maximum likelihood estimation rather than via p-value. Numerical results are provided in Supplementary Tables 1, 2.

Source data

Extended Data Fig. 2 Robustness and power of cross-trait TCSC in simulations.

(a) Bias in estimates of genetic covariance explained by the cis-genetic component of gene expression in tissue t′ (\({\omega }_{ge(t{\prime} )}\)) for causal (purples) and noncausal (grays) tissues, across n = 1,000 independent simulations per eQTL sample size (n = 100, 200, 300, 500, 1000, 1500). Light purple (resp. gray) indicates that \({G}_{t{\prime} }\) was set to the total number of true cis-heritable genes across tissues, dark purple (resp. gray) indicates that \({G}_{t{\prime} }\) was set to the number of significantly cis-heritable genes detected in each tissue. The dashed line indicates the true value of \({\omega }_{ge(t{\prime} )}\) for causal tissues. (b) Percentage of estimates of \({\omega }_{ge(t{\prime} )}\) for noncausal tissues that were significantly positive at p < 0.05. (c) Percentage of estimates of \({\omega }_{ge(t{\prime} )}\) for causal tissues that were significantly positive at p < 0.05. For panels B and C, we performed n = 1,000 independent simulations per eQTL sample size (n = 100, 200, 300, 500, 1000, 1500) and used a one-sided z-test to obtain p-values. In all panels, data are presented as mean values ± 1.96 × s.e.m. Numerical results are reported in Supplementary Table 3.

Source data

Extended Data Fig. 3 Robustness and power of TCSC regression in simulations with different values of \({{\boldsymbol{h}}}_{{\boldsymbol{ge}}({\boldsymbol{t}}{\prime} )}^{2}\).

(a) Type I error per true value of \({h}_{ge(t{\prime} )}^{2}\) in the causal tissue. False positive event is defined as \({h}_{ge(t{\prime} )}^{2}\)> 0 for noncausal tissues at p < 0.05. (b) Power to detect the causal tissue per true value of \({h}_{ge(t{\prime} )}^{2}\) in the causal tissue. A true positive event is defined as \({h}_{ge(t{\prime} )}^{2}\) > 0 for causal tissues at p < 0.05. (c) Bias on causal estimates of \({h}_{ge(t{\prime} )}^{2}\) for different true values of the causal tissue \({h}_{ge(t{\prime} )}^{2}\). Dashed lines indicate true values of \({h}_{ge(t{\prime} )}^{2}\). (d) Bias on noncausal estimates of \({h}_{ge(t{\prime} )}^{2}\) for different true values of the causal tissue \({h}_{ge(t{\prime} )}^{2}\). In all panels, we performed n = 1,000 independent simulated genetic architectures across different eQTL sample sizes (n = 100, 200, 300, 500, 1000, 1500); we used a one-sided z-test and the genomic block jackknife standard error to obtain p-values and data are presented as mean values ± 1.96 × s.e.m. The value of \({G}_{t{\prime} }\) is set to the total number of unique cis-heritable genes across all tissues.

Source data

Extended Data Fig. 4 Robustness and power of cross-trait TCSC regression in simulations with different values of \({{\boldsymbol{\omega }}}_{{\boldsymbol{ge}}({\boldsymbol{t}}{\prime} )}\).

(a) Type I error per true value of \({\omega }_{ge(t{\prime} )}\) in the causal tissue. False positive event is defined as \({\omega }_{ge(t{\prime} )}\)> 0 for noncausal tissues at p < 0.05. (b) Power to detect the causal tissue per true value of \({\omega }_{ge(t{\prime} )}\) in the causal tissue. A true positive event is defined as \({\omega }_{ge(t{\prime} )}\) > 0 for causal tissues at p < 0.05. (c) Bias on causal estimates of \({\omega }_{ge(t{\prime} )}\) for different true values of the causal tissue \({\omega }_{ge(t{\prime} )}\). Dashed lines indicate true values of \({\omega }_{ge(t{\prime} )}\). (d) Bias on noncausal estimates of \({\omega }_{ge(t{\prime} )}\) for different true values of the causal tissue \({\omega }_{ge(t{\prime} )}\). In all panels, we performed n = 1,000 independent simulated genetic architectures across different eQTL sample sizes (n = 100, 200, 300, 500, 1000, 1500); we used a one-sided z-test and the genomic block jackknife standard error to obtain p-values and data are presented as mean values ± 1.96 × s.e.m. The value of \({G}_{t{\prime} }\) is set to the total number of unique cis-heritable genes across all tissues.

Source data

Extended Data Fig. 5 Robustness and power of TCSC regression in simulations with different numbers of noncausal tissues.

(a) Type I error involving a variable number of noncausal tissues in the presence of a single causal tissue. False positive event defined as \({h}_{ge(t{\prime} )}^{2}\) > 0 for noncausal tissues at p < 0.05. Note, when there are 0 tagging tissues, there is no measurement of type I error. (b) Power to detect the causal tissue in which \({h}_{ge(t{\prime} )}^{2} > 0\) for causal tissues at p < 0.05. (c) Bias on estimates of \({h}_{ge(t{\prime} )}^{2}\) for the causal tissue, while changing the number of noncausal tissues in the model. The dashed line indicates that the true value of \({h}_{ge(t{\prime} )}^{2}\). (d) Bias on estimates of \({h}_{ge(t{\prime} )}^{2}\) for noncausal tissues, while changing the number of noncausal tissues in the model. In all panels, we performed n = 1,000 independent simulated genetic architectures across different eQTL sample sizes (n = 100, 200, 300, 500, 1000, 1500); we used a one-sided z-test and the genomic block jackknife standard error to obtain p-values and data are presented as mean values ± 1.96 × s.e.m. The value of \({G}_{t{\prime} }\) is set to the total number of unique cis-heritable genes across all tissues.

Source data

Extended Data Fig. 6 Robustness and power of cross-trait TCSC regression in simulations with different numbers of noncausal tissues.

(a) Type I error involving a variable number of noncausal tissues in the presence of a single causal tissue. False positive event defined as \({\omega }_{ge(t{\prime} )}\) > 0 for noncausal tissues at p < 0.05. Note, when there are 0 tagging tissues, there is no measurement of type I error. (b) Power to detect the causal tissue in which \({\omega }_{ge(t{\prime} )} > 0\) for causal tissues at p < 0.05. (c) Bias on estimates of \({\omega }_{ge(t{\prime} )}\) for the causal tissue, while changing the number of noncausal tissues in the model. The dashed line indicates that the true value of \({\omega }_{ge(t{\prime} )}\). (d) Bias on estimates of \({\omega }_{ge(t{\prime} )}\) for noncausal tissues, while changing the number of noncausal tissues in the model. In all panels, we performed n = 1,000 independent simulated genetic architectures across different eQTL sample sizes (n = 100, 200, 300, 500, 1000, 1500); we used a one-sided z-test and the genomic block jackknife standard error to obtain p-values and data are presented as mean values ± 1.96 × s.e.m. The value of \({G}_{t{\prime} }\) is set to the total number of unique cis-heritable genes across all tissues.

Source data

Extended Data Fig. 7 Tissue-specific contributions to disease and complex trait heritability in secondary analysis of 23 tissues, removing tissues with small eQTL sample size.

We report estimates of the proportion of disease heritability explained by the cis-genetic component of gene expression in tissue \(t{\prime}\) (\({\pi }_{t{\prime} }\)). Results are shown for tissue-trait pairs with FDR <= 10%; full boxes denote FDR of 5% or lower and partial boxes denote FDR between 5% and 10%. Tissues are ordered alphabetically. Color corresponds to \({\pi }_{t{\prime} }\), the proportion of common variant heritability causally explained by predicted gene expression in tissue \(t{\prime}\). These results are largely consistent with the analysis of 39 GTEx tissues (Fig. 4). WHRadjBMI: waist-hip-ratio adjusted for body mass index. HDL: high-density lipoprotein. DBP: diastolic blood pressure. BMI: body mass index. FEV1/FVC: forced expiratory volume in one second divided by forced vital capacity. Cereb. Cortex Ar.: cerebral cortex surface area. WBC Count: white blood cell count. RBC Count: red blood cell count. MDD: major depressive disorder. Daggers next to a tissue indicate a meta-tissue.

Source data

Extended Data Fig. 8 Tissue-specific contributions to disease and complex trait heritability in brain-specific analysis.

We separately analyzed results for 41 brain-related diseases and complex traits and 13 brain tissues. Results are shown for tissue-trait pairs with FDR <= 10%; full boxes denote FDR of 5% or lower and partial boxes denote FDR between 5% and 10%. Tissues are ordered alphabetically. Each tissue has an eQTL sample size ranging from 101 to 189 individuals. Color corresponds to \({\pi }_{t{\prime} }\), the proportion of common variant heritability causally explained by predicted gene expression in tissue t'. Caudate Vol: caudate volume. BMI: body mass index. Scz: schizophrenia. Bipolar: bipolar disorder. Brainstem Vol: brainstem volume. Cereb. Cortex Width: cerebral cortex width. ADHD: attention-deficit/hyperactivity disorder. Brainstem Vol: brainstem volume .

Source data

Extended Data Fig. 9 Comparison of disease-critical tissues identified by RTC Coloc, LDSC-SEG and TCSC for all 17 disease/traits with causal tissue-trait associations identified by TCSC.

FDR significance of trait-tissue associations across three different methods for each of 17 traits with a significant tissue found by TCSC from Fig. 3 and 7 causal tissues, plus each tissue’s most highly genetically correlated GTEx tissue. Full boxes denote FDR of 5% or lower and partial boxes denote FDR between 5% and 10%. Thicker black lines separate the causal tissue found in the primary TCSC analysis (left) from its most highly genetically correlated GTEx tissue (right), with two exceptions. First, breast tissue was the most highly genetically correlated tissue for two causal tissues, adipose subcutaneous and thyroid; therefore, these three tissues appear as a trio. Second, the aorta artery and tibial artery are each other’s most highly genetically correlated tissue and both are a causal tissue different traits by TCSC. (a) RTC Coloc (Ongen 2017 Nat Genet), (b) LDSC-SEG (Finucane 2018 Nat Genet), (c) TCSC. Per-trait FDR in panels A and C, FDR across traits and tissues in panel B. WHRadjBMI: waist-hip-ratio conditional on body mass index. HDL: high-density lipoprotein. DBP: diastolic blood pressure. BMI: body mass index. FEV1/FVC: forced expiratory volume in one second divided by forced vital capacity. Cereb. Cortex Ar.: cerebral cortex surface area. AST: aspartate aminotransferase. LDL: low-density lipoprotein. WBC: white blood cell count. MDD: major depressive disorder. Daggers next to a tissue indicate a meta-tissue. For BMI, fecundity, and cereb. cortex ar., LDSC-SEG brain-specific analysis results are used.

Source data

Extended Data Fig. 10 Comparison of disease-critical tissues identified by RTC Coloc, LDSC-SEG and TCSC for brain-specific analysis.

ac, Brain-specific tissue-trait pairs identified by three methods: RTC Coloc (a), LDSC-SEG (b) and TCSC (c). Full boxes denote FDR of 5% or lower and partial boxes denote FDR between 5% and 10%. Each tissue has an eQTL sample size ranging from 100 to 189 individuals. Additionally, we include each tissue’s most highly genetically correlated GTEx tissue. Thicker black lines separate the causal tissue found in the primary TCSC analysis (left) from its most highly genetically correlated GTEx tissue (right), with one exception: brain accumbens, caudate, putamen, hippocampus, and amygdala are all highly genetically correlated, with no pairs of exclusively high genetic correlation. Caudate Vol: caudate volume. BMI: body mass index. Scz: schizophrenia. Bipolar: bipolar disorder. Brainstem Vol: brainstem volume. Cereb. Cortex Width: cerebral cortex width. ADHD: attention-deficit/hyperactivity disorder. Brainstem Vol: brainstem volume.

Source data

Supplementary information

Supplementary Information

Supplementary Note, Figs. 1–22 and table legends.

Reporting Summary

Supplementary Tables 1–21

Numerical results.

Source data

Source Data

Source data for Figs. 2–5 and Extended Data Figs. 1–10.

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Amariuta, T., Siewert-Rocks, K. & Price, A.L. Modeling tissue co-regulation estimates tissue-specific contributions to disease. Nat Genet 55, 1503–1511 (2023). https://doi.org/10.1038/s41588-023-01474-z

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41588-023-01474-z

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