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.

Genomic atlas of the human plasma proteome

Abstract

Although plasma proteins have important roles in biological processes and are the direct targets of many drugs, the genetic factors that control inter-individual variation in plasma protein levels are not well understood. Here we characterize the genetic architecture of the human plasma proteome in healthy blood donors from the INTERVAL study. We identify 1,927 genetic associations with 1,478 proteins, a fourfold increase on existing knowledge, including trans associations for 1,104 proteins. To understand the consequences of perturbations in plasma protein levels, we apply an integrated approach that links genetic variation with biological pathway, disease, and drug databases. We show that protein quantitative trait loci overlap with gene expression quantitative trait loci, as well as with disease-associated loci, and find evidence that protein biomarkers have causal roles in disease using Mendelian randomization analysis. By linking genetic factors to diseases via specific proteins, our analyses highlight potential therapeutic targets, opportunities for matching existing drugs with new disease indications, and potential safety concerns for drugs under development.

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: The genetic architecture of plasma protein levels.
Fig. 2: Missense variant rs28929474:T in SERPINA1 is a trans pQTL hotspot.
Fig. 3: trans pQTL for BLIMP1 at an inflammatory bowel disease (IBD) associated missense variant (rs3197999:A) in MST1.
Fig. 4: Proteinase-3, SERPINA1 and vasculitis.
Fig. 5: Evaluation of causal role of proteins in disease.

Similar content being viewed by others

References

  1. Albert, F. W. & Kruglyak, L. The role of regulatory variation in complex traits and disease. Nat. Rev. Genet. 16, 197–212 (2015).

    Article  PubMed  CAS  Google Scholar 

  2. Liu, Y. et al. Quantitative variability of 342 plasma proteins in a human twin population. Mol. Syst. Biol. 11, 786 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  3. Suhre, K. et al. Connecting genetic risk to disease end points through the human blood plasma proteome. Nat. Commun. 8, 14357 (2017).

    Article  ADS  PubMed  PubMed Central  CAS  Google Scholar 

  4. Yao, C. et al. Genome-wide association study of plasma proteins identifies putatively causal genes, proteins, and pathways for cardiovascular disease. Preprint at https://www.biorxiv.org/content/early/2017/05/12/136523 (2017).

  5. de Vries, P. S. et al. Whole-genome sequencing study of serum peptide levels: the Atherosclerosis Risk in Communities study. Hum. Mol. Genet. 26, 3442–3450 (2017).

    Article  PubMed  CAS  PubMed Central  Google Scholar 

  6. Wu, L. et al. Variation and genetic control of protein abundance in humans. Nature 499, 79–82 (2013).

    Article  ADS  PubMed  PubMed Central  CAS  Google Scholar 

  7. Battle, A. et al. Impact of regulatory variation from RNA to protein. Science 347, 664–667 (2015).

    Article  ADS  PubMed  CAS  Google Scholar 

  8. Rohloff, J. C. et al. Nucleic acid ligands with protein-like side chains: modified aptamers and their use as diagnostic and therapeutic agents. Mol. Ther. Nucleic Acids 3, e201 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  9. Di Angelantonio, E. et al. Efficiency and safety of varying the frequency of whole blood donation (INTERVAL): a randomised trial of 45 000 donors. Lancet 390, 2360–2371 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  10. 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).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  11. Burgess, S., Scott, R. A., Timpson, N. J., Davey Smith, G. & Thompson, S. G. Using published data in Mendelian randomization: a blueprint for efficient identification of causal risk factors. Eur. J. Epidemiol. 30, 543–552 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  12. Stranger, B. E. et al. Patterns of cis regulatory variation in diverse human populations. PLoS Genet. 8, e1002639 (2012).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  13. Lundberg, M., Eriksson, A., Tran, B., Assarsson, E. & Fredriksson, S. Homogeneous antibody-based proximity extension assays provide sensitive and specific detection of low-abundant proteins in human blood. Nucleic Acids Res. 39, e102 (2011).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  14. Walter, K. et al. The UK10K project identifies rare variants in health and disease. Nature 526, 82–90 (2015).

    Article  ADS  PubMed  CAS  Google Scholar 

  15. Westra, H.-J. et al. Systematic identification of trans eQTLs as putative drivers of known disease associations. Nat. Genet. 45, 1238–1243 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  16. Lappalainen, T. et al. Transcriptome and genome sequencing uncovers functional variation in humans. Nature 501, 506–511 (2013).

    Article  ADS  PubMed  PubMed Central  CAS  Google Scholar 

  17. Schadt, E. E. et al. Mapping the genetic architecture of gene expression in human liver. PLoS Biol. 6, e107 (2008).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  18. Zeller, T. et al. Genetics and beyond—the transcriptome of human monocytes and disease susceptibility. PLoS ONE 5, e10693 (2010).

    Article  ADS  PubMed  PubMed Central  CAS  Google Scholar 

  19. Nai, A. et al. TMPRSS6 rs855791 modulates hepcidin transcription in vitro and serum hepcidin levels in normal individuals. Blood 118, 4459–4462 (2011).

    Article  PubMed  CAS  Google Scholar 

  20. Carrasquillo, M. M. et al. Genome-wide screen identifies rs646776 near sortilin as a regulator of progranulin levels in human plasma. Am. J. Hum. Genet. 87, 890–897 (2010).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  21. Gooptu, B., Dickens, J. A. & Lomas, D. A. The molecular and cellular pathology of α1-antitrypsin deficiency. Trends Mol. Med. 20, 116–127 (2014).

    Article  PubMed  CAS  Google Scholar 

  22. Stacey, D. et al. ProGeM: A framework for the prioritisation of candidate causal genes at molecular quantitative trait loci. https://doi.org/10.1101/230094 (2017).

  23. 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).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  24. Di Narzo, A. F. et al. High-throughput characterization of blood serum proteomics of IBD patients with respect to aging and genetic factors. PLoS Genet. 13, e1006565 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  25. Huang, H. et al. Fine-mapping inflammatory bowel disease loci to single-variant resolution. Nature 547, 173–178 (2017).

    Article  ADS  PubMed  PubMed Central  CAS  Google Scholar 

  26. Lyons, P. A. et al. Genetically distinct subsets within ANCA-associated vasculitis. N. Engl. J. Med. 367, 214–223 (2012).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  27. Merkel, P. A. et al. Identification of functional and expression polymorphisms associated with risk for anti-neutrophil cytoplasmic autoantibody-associated vasculitis. Arthritis Rheumatol. 69, 1054–1066 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  28. Battle, A., Brown, C. D., Engelhardt, B. E. & Montgomery, S. B. Genetic effects on gene expression across human tissues. Nature 550, 204–213 (2017).

    Article  ADS  PubMed  Google Scholar 

  29. Grönberg, H. et al. Prostate cancer screening in men aged 50-69 years (STHLM3): a prospective population-based diagnostic study. Lancet Oncol. 16, 1667–1676 (2015).

    Article  PubMed  Google Scholar 

  30. Eeles, R. A. et al. Multiple newly identified loci associated with prostate cancer susceptibility. Nat. Genet. 40, 316–321 (2008).

    Article  PubMed  CAS  Google Scholar 

  31. Paternoster, L. et al. Multi-ancestry genome-wide association study of 21,000 cases and 95,000 controls identifies new risk loci for atopic dermatitis. Nat. Genet. 47, 1449–1456 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  32. Dahl, R. et al. Effects of an oral MMP-9 and -12 inhibitor, AZD1236, on biomarkers in moderate/severe COPD: a randomised controlled trial. Pulm. Pharmacol. Ther. 25, 169–177 (2012).

    Article  PubMed  CAS  Google Scholar 

  33. Ganz, P. et al. Development and validation of a protein-based risk score for cardiovascular outcomes among patients with stable coronary heart disease. J. Am. Med. Assoc. 315, 2532–2541 (2016).

    Article  CAS  Google Scholar 

  34. Traylor, M. et al. A novel MMP12 locus is associated with large artery atherosclerotic stroke using a genome-wide age-at-onset informed approach. PLoS Genet. 10, e1004469 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  35. Nelson, M. R. et al. The support of human genetic evidence for approved drug indications. Nat. Genet. 47, 856–860 (2015).

    Article  PubMed  CAS  Google Scholar 

  36. Albagha, O. M. E. et al. Genome-wide association study identifies variants at CSF1, OPTN and TNFRSF11A as genetic risk factors for Paget’s disease of bone. Nat. Genet. 42, 520–524 (2010).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  37. Schwarz, P., Rasmussen, A. Q., Kvist, T. M., Andersen, U. B. & Jørgensen, N. R. Paget’s disease of the bone after treatment with Denosumab: a case report. Bone 50, 1023–1025 (2012).

    Article  PubMed  Google Scholar 

  38. Moore, C. et al. The INTERVAL trial to determine whether intervals between blood donations can be safely and acceptably decreased to optimise blood supply: study protocol for a randomised controlled trial. Trials 15, 363 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  39. Gold, L. et al. Aptamer-based multiplexed proteomic technology for biomarker discovery. PLoS ONE 5, e15004 (2010).

    Article  ADS  PubMed  PubMed Central  CAS  Google Scholar 

  40. Sattlecker, M. et al. Alzheimer’s disease biomarker discovery using SOMAscan multiplexed protein technology. Alzheimers Dement. 10, 724–734 (2014).

    Article  PubMed  Google Scholar 

  41. McLaren, W. et al. Deriving the consequences of genomic variants with the Ensembl API and SNP Effect Predictor. Bioinformatics 26, 2069–2070 (2010).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  42. UniProt Consortium. UniProt: a hub for protein information. Nucleic Acids Res. 43, D204–D212 (2015).

    Article  CAS  Google Scholar 

  43. Ashburner, M. et al.; The Gene Ontology Consortium. Gene ontology: tool for the unification of biology. Nat. Genet. 25, 25–29 (2000).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  44. Menni, C. et al. Circulating proteomic signatures of chronological age. J. Gerontol. A Biol. Sci. Med. Sci. 70, 809–816 (2015).

    Article  PubMed  CAS  Google Scholar 

  45. Ngo, D. et al. Aptamer-based proteomic profiling reveals novel candidate biomarkers and pathways in cardiovascular disease. Circulation 134, 270–285 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  46. Chang, C. C. et al. Second-generation PLINK: rising to the challenge of larger and richer datasets. Gigascience 4, 7 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  47. 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).

    Article  PubMed  CAS  Google Scholar 

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

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  49. 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  PubMed  PubMed Central  CAS  Google Scholar 

  50. Welter, D. et al. The NHGRI GWAS Catalog, a curated resource of SNP-trait associations. Nucleic Acids Res. 42, D1001–D1006 (2014).

    Article  PubMed  CAS  Google Scholar 

  51. Malone, J. et al. Modeling sample variables with an Experimental Factor Ontology. Bioinformatics 26, 1112–1118 (2010).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  52. Javierre, B. M. et al. Lineage-specific genome architecture links enhancers and non-coding disease variants to target gene promoters. Cell 167, 1369–1384. e19 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  53. Lawrence, M. et al. Software for computing and annotating genomic ranges. PLOS Comput. Biol. 9, e1003118 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  54. Amberger, J. S., Bocchini, C. A., Schiettecatte, F., Scott, A. F. & Hamosh, A. OMIM.org: Online Mendelian Inheritance in Man (OMIM®), an online catalog of human genes and genetic disorders. Nucleic Acids Res. 43, D789–D798 (2015).

    Article  PubMed  CAS  Google Scholar 

  55. Kanehisa, M., Sato, Y., Kawashima, M., Furumichi, M. & Tanabe, M. KEGG as a reference resource for gene and protein annotation. Nucleic Acids Res. 44, D457–D462 (2016).

    Article  PubMed  CAS  Google Scholar 

  56. Szklarczyk, D. et al. STRINGv10: protein-protein interaction networks, integrated over the tree of life. Nucleic Acids Res. 43, D447–D452 (2015).

    Article  PubMed  CAS  Google Scholar 

  57. Smith, R. N. et al. InterMine: a flexible data warehouse system for the integration and analysis of heterogeneous biological data. Bioinformatics 28, 3163–3165 (2012).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  58. Franceschini, A. et al. STRINGv9.1: protein–protein interaction networks, with increased coverage and integration. Nucleic Acids Res. 41, D808–D815 (2013).

    Article  PubMed  CAS  Google Scholar 

  59. Lek, M. et al. Analysis of protein-coding genetic variation in 60,706 humans. Nature 536, 285–291 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  60. Iotchkova, V. et al. GARFIELD—GWAS Analysis of Regulatory or Functional Information Enrichment with LD correction. Preprint at https://www.biorxiv.org/content/early/2016/11/07/085738 (2016).

  61. Staley, J. R. et al. PhenoScanner: a database of human genotype–phenotype associations. Bioinformatics 32, 3207–3209 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

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

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  63. Hingorani, A. & Humphries, S. Nature’s randomised trials. Lancet 366, 1906–1908 (2005).

    Article  PubMed  Google Scholar 

  64. Nikpay, M. et al. A comprehensive 1,000 Genomes-based genome-wide association meta-analysis of coronary artery disease. Nat. Genet. 47, 1121–1130 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  65. Burgess, S., Butterworth, A. & Thompson, S. G. Mendelian randomization analysis with multiple genetic variants using summarized data. Genet. Epidemiol. 37, 658–665 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  66. Burgess, S., Dudbridge, F. & Thompson, S. G. Combining information on multiple instrumental variables in Mendelian randomization: comparison of allele score and summarized data methods. Stat. Med. 35, 1880–1906 (2016).

    Article  MathSciNet  PubMed  Google Scholar 

  67. Burgess, S. & Thompson, S. G. Multivariable Mendelian randomization: the use of pleiotropic genetic variants to estimate causal effects. Am. J. Epidemiol. 181, 251–260 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  68. Burgess, S., Dudbridge, F. & Thompson, S. G. Re: “Multivariable Mendelian randomization: the use of pleiotropic genetic variants to estimate causal effects”. Am. J. Epidemiol. 181, 290–291 (2015).

    Article  PubMed  Google Scholar 

Download references

Acknowledgements

A. Day-Williams, J. McElwee, D. Diogo, W. Astle, E. Di Angelantonio, E. Birney, A. Richard, J. Mason and M. Inouye commented on the manuscript, and M. Sharp helped with mapping drug indications to GWAS traits. We thank INTERVAL study participants; staff at recruiting NHSBT blood donation centres; and the INTERVAL Study Co-ordination team, Operations Team (led by R. Houghton and C. Moore) and Data Management Team (led by M. Walker). Funding sources are listed in the Supplementary Information.

Reviewer information

Nature thanks T. Lappalainen, M. McCarthy and the other anonymous reviewer(s) for their contribution to the peer review of this work.

Author information

Authors and Affiliations

Authors

Contributions

Conceptualization and experimental design: J.D., A.S.B., B.B.S., H.R., R.M.P.; methodology: B.B.S., A.S.B., J.C.M., J.E.P., H.R., S.B.; conducted experimental work: N.J., S.K.W., E.S.Z.; analysis: B.B.S., J.C.M., J.E.P., D.S., J.B., J.R.S., T.J., E.P., P.S., C.O.-W., M.A.K., S.K.W., A.C., N.B., S.L.S.; contributed reagents, materials, protocols or analysis tools: N.J., S.K.W., E.S.Z., J.B., M.A.K., J.R.S., B.P.P.; supervision: A.S.B., H.R., J.D., R.M.P., C.S.F., D.S.P., A.M.W.; writing: A.S.B., J.E.P., B.B.S., J.C.M., H.R., J.D., J.A.T., N.S., K.S.; creation of the INTERVAL BioResource: J.R.B., D.J.R.,W.H.O., N.W.M., J.D.; funding: N.W.M., J.R.B., D.J.R., W.H.O., H.R., R.M.P., J.D.; all authors critically reviewed the manuscript.

Corresponding authors

Correspondence to John Danesh or Adam S. Butterworth.

Ethics declarations

Competing interests

The authors declare the following competing interests: A.C., CSF-Merck employee; N.J., S.K.W., SomaLogic Inc employees and stakeholders; E.S.Z., SomaLogic Inc employee; J.C.M., R.M.P., Merck employees during this study, now Celgene employees; H.R., Merck employee during this study; J.E.P., travel and accommodation expenses and hospitality from Olink to speak at Olink-sponsored academic meetings; A.S.B., grants from Merck, Pfizer, Novartis, Biogen and Bioverativ and personal fees from Novartis; J.D., sits on the Novartis Cardiovascular and Metabolic Advisory Board, had grant support from Novartis.

Additional information

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

Extended data figures and tables

Extended Data Fig. 1

Flowchart of sample processing and quality control stages for proteomic and genetic measurements before genetic analyses.

Extended Data Fig. 2 Examples of protein targets for which the SOMAmer is highly specific.

SDS–PAGE with Alexa-647-labelled proteins captured by the IL1RL2 SOMAmer (a) or GP1BA SOMAmer (b). For each protein target, the protein captured by the SOMAmer is compared to the standard. The cognate targets are the only ones with protein visible in the capture lanes, whereas the proteins homologous to the target proteins show no evidence of binding. These experiments were performed once. MW markers, molecular weight markers.

Extended Data Fig. 3 Evidence for the reliability of protein measurements made using the SOMAscan assay.

a, Distribution of coefficients of variation of all proteins on the SOMAscan assay in each subcohort. b, Spearman’s correlations for all proteins passing QC derived from contemporaneous assay of baseline and two-year samples from 60 participants. c, Scatterplot of pQTL effect size estimates from SOMAscan versus Olink showing all 163 pQTLs tested (top) and the 106 that replicated (bottom). r is Pearson’s correlation coefficient. d, Distribution of inflation factors across proteins that underwent genome-wide association testing, stratified by subcohort and allele frequency (MAF ≥ 5%, MAF < 5%).

Extended Data Fig. 4 The WFIKKN2 region is a trans pQTL for GDF11/8 plasma levels.

a, Regional association plots of the trans pQTL (sentinel variant rs11079936) for GDF11/8 before and after adjusting for levels of WFIKKN2 (upper panels), and the WFIKKN2 cis pQTL after adjusting for GDF11/8 levels (bottom panel). A similar pattern of association for WFIKKN2 was seen before GDF11/8 adjustment (not shown). b, Attenuation of the GDF11/8 trans pQTL upon adjustment for plasma levels of the cis protein WFIKKN2.

Extended Data Fig. 5 Genetic architecture of the pQTLs.

pQTL mapping in n = 3,301 individuals. a, Distribution of the predicted consequences of the sentinel pQTL variants compared to matched permuted null sets of variants, stratified by cis and trans. Asterisks indicate empirical enrichment using a permutation test (10,000 permuted sets of non-associated variants) at a Bonferroni-corrected significance value (P < 0.005). Bar height represents the mean proportion of variants within each class and error bars reflect one standard deviation from the mean. b, Number of proteins associated (P < 1.5 × 10−11) with each sentinel variant across the genome.

Extended Data Fig. 6 Enrichment of pQTLs at DNase I hypersensitive sites by tissue or cell type.

Circle shows enrichment for DNase I hypersensitive sites (‘hotspots’) for each of 55 tissues (183 cell types) available from the ENCODE and Roadmap Epigenomics projects, with tissues or cell types clustered and coloured by anatomical grouping. Some tissues have multiple values due to availability of multiple cell types or multiple tests per cell type. Radial lines show fold-enrichment, while dots around the inside edge of the circle denote statistically significant enrichment at a Bonferroni-corrected significant threshold P < 5 × 10−5. Enrichment testing performed using GARFIELD (which tests enrichment against permuted sets of variants matched for MAF, distance to TSS and LD). pQTL data from n = 3,301 individuals.

Extended Data Fig. 7 Scheme outlining the combined ‘bottom-up’ and ‘top-down’ process used for candidate gene annotation of trans pQTL regions.

See Methods. GbA, guilt-by-association; KEGG, Kyoto Encyclopedia of Genes and Genomes; OMIM, Online Mendelian Inheritance in Man; STRINGdb, STRING database.

Extended Data Fig. 8 Follow-up of PR3 SOMAmers.

These experiments were repeated three times independently with similar results. a, SOMAmer pulldowns with purified PR3, A1AT, and PR3–A1AT complex. SOMAmer PRTN3.3514.49.2 enriched the PR3–A1AT complex to a much greater degree than free PR3. Conversely, SOMAmer PRTN3.13720.95.3 enriched free PR3 to a greater degree than the PR3–A1AT complex. b, Solution affinity of PRTN3.3514.49.2 and PRTN3.13720.95.3 for PR3, A1AT, and the PR3–A1AT complex. SOMAmer PRTN3.3514.49.2 has a higher affinity for the PR3–A1AT complex than for free PR3. SOMAmer PRTN3.13720.95.3, on the other hand, has a higher affinity for free PR3 than SOMAmer PRTN3.3514.49.2. c, Competitive binding of SOMAmers PRTN3.13720.95.3 and PRTN3.3514.49.2 to PR3. A limiting amount of radiolabelled PRTN3.13720.95.3 was incubated with 1 nM proteinase-3 and a titration of either cold PRTN3.13720.95.3 or cold PRTN3.3514.49.2.

Extended Data Fig. 9

Comparison between a randomized controlled trial and Mendelian randomization to assess the causal effect of changes in protein biomarker levels on disease risk.

Extended Data Fig. 10 Characterization of protein targets measured using the SOMAscan assay.

a, Compartment distribution with annotations of all proteins in the Human Protein Atlas for comparison. b, GO molecular functions.

Supplementary information

Supplementary Information

This file contains funding details, full Supplementary Table Legends, Supplementary Notes and Supplementary References

Reporting Summary

Supplementary Figure 1

A three-dimensional interactive plot of sentinel variant-protein associations (red-cis, blue-trans). X-axis (“pQTL position”) represents position of the sentinel variant along chromosomes 1-22. Y-axis (“Protein position”) represents the start position of the gene encoding the protein. Z-axis represents the –log10(p) of the association. Additional details can be viewed when hovering over the points. Clicking on cis/trans in the legend toggles display of points by cis/trans. Additional viewing controls are available at the top right of the window. For clarity, associations with p<10-300 (diamonds) are plotted at -log10(p)=300. The plot is generated using “plotly” R package v4.5.6 (Plotly Technologies Inc., Montréal, Canada)

Supplementary Tables

Supplementary Tables 1-21 – see Supplementary Information file for full descriptions

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Sun, B.B., Maranville, J.C., Peters, J.E. et al. Genomic atlas of the human plasma proteome. Nature 558, 73–79 (2018). https://doi.org/10.1038/s41586-018-0175-2

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41586-018-0175-2

This article is cited by

Comments

By submitting a comment you agree to abide by our Terms and Community Guidelines. If you find something abusive or that does not comply with our terms or guidelines please flag it as inappropriate.

Search

Quick links

Nature Briefing: Translational Research

Sign up for the Nature Briefing: Translational Research newsletter — top stories in biotechnology, drug discovery and pharma.

Get what matters in translational research, free to your inbox weekly. Sign up for Nature Briefing: Translational Research