High-Throughput Characterization of Blood Serum Proteomics of IBD Patients with Respect to Aging and Genetic Factors

Antonio F Di Narzo, Shannon E Telesco, Carrie Brodmerkel, Carmen Argmann, Lauren A Peters, Katherine Li, Brian Kidd, Joel Dudley, Judy Cho, Eric E Schadt, Andrew Kasarskis, Radu Dobrin, Ke Hao, Antonio F Di Narzo, Shannon E Telesco, Carrie Brodmerkel, Carmen Argmann, Lauren A Peters, Katherine Li, Brian Kidd, Joel Dudley, Judy Cho, Eric E Schadt, Andrew Kasarskis, Radu Dobrin, Ke Hao

Abstract

To date, no large scale, systematic description of the blood serum proteome has been performed in inflammatory bowel disease (IBD) patients. By using microarray technology, a more complete description of the blood proteome of IBD patients is feasible. It may help to achieve a better understanding of the disease. We analyzed blood serum profiles of 1128 proteins in IBD patients of European descent (84 Crohn's Disease (CD) subjects and 88 Ulcerative Colitis (UC) subjects) as well as 15 healthy control subjects, and linked protein variability to patient age (all cohorts) and genetic components (genotype data generated from CD patients). We discovered new, previously unreported aging-associated proteomic traits (such as serum Albumin level), confirmed previously reported results from different tissues (i.e., upregulation of APOE with aging), and found loss of regulation of MMP7 in CD patients. In carrying out a genome wide genotype-protein association study (proteomic Quantitative Trait Loci, pQTL) within the CD patients, we identified 41 distinct proteomic traits influenced by cis pQTLs (underlying SNPs are referred to as pSNPs). Significant overlaps between pQTLs and cis eQTLs corresponding to the same gene were observed and in some cases the QTL were related to inflammatory disease susceptibility. Importantly, we discovered that serum protein levels of MST1 (Macrophage Stimulating 1) were regulated by SNP rs3197999 (p = 5.96E-10, FDR<5%), an accepted GWAS locus for IBD. Filling the knowledge gap of molecular mechanisms between GWAS hits and disease susceptibility requires systematically dissecting the impact of the locus at the cell, mRNA expression, and protein levels. The technology and analysis tools that are now available for large-scale molecular studies can elucidate how alterations in the proteome driven by genetic polymorphisms cause or provide protection against disease. Herein, we demonstrated this directly by integrating proteomic and pQTLs with existing GWAS, mRNA expression, and eQTL datasets to provide insights into the biological processes underlying IBD and pinpoint causal genetic variants along with their downstream molecular consequences.

Trial registration: ClinicalTrials.gov NCT00771667.

Conflict of interest statement

We have read the journal's policy and the authors of this manuscript have the following competing interests. The following authors: SET, CB, KL and RD are paid employees of Janssen R&D LLC. The work is partially supported by Janssen R&D LLC.

Figures

Fig 1. Heatmap of top significant age-related…
Fig 1. Heatmap of top significant age-related proteins.
Included in the heatmap are all microarray probes with pvalue ≤ 1E-4 in at least one cohort. The UC, CD and NC cells are color-coded according to the Wald t-test of the age coefficient of the protein levels in each cohort, with the t test further reported in each cell. The last 2 columns report previously known association of gene mRNA levels with age within different tissues. Shades of green indicate increase of protein or mRNA level with aging; shades of red indicate decrease of protein or mRNA level with aging; the white color indicates lack of evidence in either direction.
Fig 2. Increase of Albumin levels with…
Fig 2. Increase of Albumin levels with aging in CD and UC.
Scatterplot of the Albumin protein level vs patients age, separately for UC patients (left panel) and CD patients (baseline data only is displayed, right panel). Age in years on the horizontal axis; mean-centered and adjusted log2-protein expression on the vertical axis (adjusted for sex and plateID).
Fig 3. Variation of APOE levels with…
Fig 3. Variation of APOE levels with aging in CD, UC and NC subjects.
Forest plot of the estimated log2-FC of APOE protein levels (probe SL000276) per 10 years increase in age, with 95% confidence intervals, as obtained from the differential expression analysis performed separately in Crohn’s Disease (CD), Ulcerative Colitis (UC) and Normal Controls (NC) subjects. Estimated log2-FCs and 95% Confidence Intervals are further reported on the right.
Fig 4. Enrichment for GWAS signals in…
Fig 4. Enrichment for GWAS signals in blood protein-QTLs of CD patients.
Expected uniform -log10(relative rank) of the protein-SNPs (nominal pvalue ≤ 1E-5) within the full GWAS SNPs list on the horizontal axis; observed–log10(relative rank) on the vertical axis. CD: Crohn’s Disease; UC: Ulcerative Colitis; BMI: Body Mass Index; SCZ: Schizofrenia; Stroke: Ischemic Stroke; T2D: Type-2 Diabetes. References for all the studies are reported in the Methods section.
Fig 5. Locuszoom plot of MST1, CD…
Fig 5. Locuszoom plot of MST1, CD and UC association pvalues around the MST1 gene.
It is worth noting that our proteomics platform has 4 probes in this chromosomal region, targeting 4 different proteins: IMPDH2 (probe SL010928), MST1 (probe SL005202), MST1R (probe SL004637) and MAPKAPK3 (probe SL004765). Of these, MST1 is most significantly associated with the IBD GWAS SNP in this locus (S3 Fig), and the association pattern was highly consistent with the CD and UC GWAS peaks (Fig 5).

References

    1. Gold L, Walker JJ, Wilcox SK, Williams S. Advances in human proteomics at high scale with the SOMAscan proteomics platform. New biotechnology. 2012;29(5):543–9. 10.1016/j.nbt.2011.11.016
    1. Hensley P. SOMAmers and SOMAscan–A Protein Biomarker Discovery Platform for Rapid Analysis of Sample Collections From Bench Top to the Clinic. Journal of biomolecular techniques: JBT. 2013;24(Suppl):S5.
    1. Sattlecker M, Kiddle SJ, Newhouse S, Proitsi P, Nelson S, Williams S, et al. Alzheimer's disease biomarker discovery using SOMAscan multiplexed protein technology. Alzheimer's & dementia: the journal of the Alzheimer's Association. 2014;10(6):724–34. Epub 2014/04/29.
    1. Menni C, Kiddle SJ, Mangino M, Viñuela A, Psatha M, Steves C, et al. Circulating proteomic signatures of chronological age. The Journals of Gerontology Series A: Biological Sciences and Medical Sciences. 2014:glu121.
    1. Horvath S. DNA methylation age of human tissues and cell types. Genome biology. 2013;14(10):1–20.
    1. Yang J, Huang T, Petralia F, Long Q, Zhang B, Argmann C, et al. Synchronized age-related gene expression changes across multiple tissues in human and the link to complex diseases. Scientific reports. 2015;5.
    1. Rodwell GE, Sonu R, Zahn JM, Lund J, Wilhelmy J, Wang L, et al. A transcriptional profile of aging in the human kidney. PLoS biology. 2004;2(12):e427 Epub 2004/11/25. 10.1371/journal.pbio.0020427
    1. Kayo T, Allison DB, Weindruch R, Prolla TA. Influences of aging and caloric restriction on the transcriptional profile of skeletal muscle from rhesus monkeys. Proceedings of the National Academy of Sciences of the United States of America. 2001;98(9):5093–8. Epub 2001/04/20. 10.1073/pnas.081061898
    1. Giau VV, Bagyinszky E, An SS, Kim SY. Role of apolipoprotein E in neurodegenerative diseases. Neuropsychiatric disease and treatment. 2015;11:1723–37. Epub 2015/07/28. 10.2147/NDT.S84266
    1. Salzman NH. Paneth cell defensins and the regulation of the microbiome: detente at mucosal surfaces. Gut microbes. 2010;1(6):401–6. Epub 2011/04/07. 10.4161/gmic.1.6.14076
    1. Wen X, Luca F, ique-Regi R. Cross-population Joint Analysis of eQTLs: Fine Mapping and Functional Annotation. 2015.
    1. Westra HJ, Peters MJ, Esko T, Yaghootkar H, Schurmann C, Kettunen J, et al. Systematic identification of trans eQTLs as putative drivers of known disease associations. Nature genetics. 2013;45(10):1238–43. Epub 2013/09/10. 10.1038/ng.2756
    1. Jostins L, Ripke S, Weersma RK, Duerr RH, McGovern DP, Hui KY, et al. Host-microbe interactions have shaped the genetic architecture of inflammatory bowel disease. Nature. 2012;491(7422):119–24. Epub 2012/11/07. 10.1038/nature11582
    1. Locke AE, Kahali B, Berndt SI, Justice AE, Pers TH, Day FR, et al. Genetic studies of body mass index yield new insights for obesity biology. Nature. 2015;518(7538):197–206. Epub 2015/02/13. 10.1038/nature14177
    1. Schizophrenia Working Group of the Psychiatric Genomics C. Biological insights from 108 schizophrenia-associated genetic loci. Nature. 2014;511(7510):421–7. . 10.1038/nature13595
    1. Traylor M, Farrall M, Holliday EG, Sudlow C, Hopewell JC, Cheng YC, et al. Genetic risk factors for ischaemic stroke and its subtypes (the METASTROKE collaboration): a meta-analysis of genome-wide association studies. The Lancet Neurology. 2012;11(11):951–62. Epub 2012/10/09. 10.1016/S1474-4422(12)70234-X
    1. Morris AP, Voight BF, Teslovich TM, Ferreira T, Segre AV, Steinthorsdottir V, et al. Large-scale association analysis provides insights into the genetic architecture and pathophysiology of type 2 diabetes. Nature genetics. 2012;44(9):981–90. Epub 2012/08/14. 10.1038/ng.2383
    1. Saad M, Lesage S, Saint-Pierre A, Corvol JC, Zelenika D, Lambert JC, et al. Genome-wide association study confirms BST1 and suggests a locus on 12q24 as the risk loci for Parkinson's disease in the European population. Human molecular genetics. 2011;20(3):615–27. Epub 2010/11/19. 10.1093/hmg/ddq497
    1. Chen ML, Lin CH, Lee MJ, Wu RM. BST1 rs11724635 interacts with environmental factors to increase the risk of Parkinson's disease in a Taiwanese population. Parkinsonism & related disorders. 2014;20(3):280–3. Epub 2013/12/18.
    1. Satake W, Nakabayashi Y, Mizuta I, Hirota Y, Ito C, Kubo M, et al. Genome-wide association study identifies common variants at four loci as genetic risk factors for Parkinson's disease. Nature genetics. 2009;41(12):1303–7. Epub 2009/11/17. 10.1038/ng.485
    1. Simon-Sanchez J, van Hilten JJ, van de Warrenburg B, Post B, Berendse HW, Arepalli S, et al. Genome-wide association study confirms extant PD risk loci among the Dutch. European journal of human genetics: EJHG. 2011;19(6):655–61. Epub 2011/01/21. 10.1038/ejhg.2010.254
    1. Nalls MA, Pankratz N, Lill CM, Do CB, Hernandez DG, Saad M, et al. Large-scale meta-analysis of genome-wide association data identifies six new risk loci for Parkinson's disease. Nature genetics. 2014;46(9):989–93. Epub 2014/07/30. 10.1038/ng.3043
    1. Lavagno L, Ferrero E, Ortolan E, Malavasi F, Funaro A. CD157 is part of a supramolecular complex with CD11b/CD18 on the human neutrophil cell surface. Journal of biological regulators and homeostatic agents. 2007;21(1–2):5–11. Epub 2008/01/24.
    1. Funaro A, Ortolan E, Ferranti B, Gargiulo L, Notaro R, Luzzatto L, et al. CD157 is an important mediator of neutrophil adhesion and migration. Blood. 2004;104(13):4269–78. Epub 2004/08/26. 10.1182/blood-2004-06-2129
    1. Davies RW, Wells GA, Stewart AF, Erdmann J, Shah SH, Ferguson JF, et al. A genome-wide association study for coronary artery disease identifies a novel susceptibility locus in the major histocompatibility complex. Circulation Cardiovascular genetics. 2012;5(2):217–25. Epub 2012/02/10. 10.1161/CIRCGENETICS.111.961243
    1. Wilk JB, Walter RE, Laramie JM, Gottlieb DJ, O'Connor GT. Framingham Heart Study genome-wide association: results for pulmonary function measures. BMC medical genetics. 2007;8 Suppl 1:S8. Epub 2007/10/16.
    1. Ferreira MA, Matheson MC, Duffy DL, Marks GB, Hui J, Le Souef P, et al. Identification of IL6R and chromosome 11q13.5 as risk loci for asthma. Lancet (London, England). 2011;378(9795):1006–14. Epub 2011/09/13.
    1. Dehghan A, Dupuis J, Barbalic M, Bis JC, Eiriksdottir G, Lu C, et al. Meta-analysis of genome-wide association studies in >80 000 subjects identifies multiple loci for C-reactive protein levels. Circulation. 2011;123(7):731–8. Epub 2011/02/09. 10.1161/CIRCULATIONAHA.110.948570
    1. Okada Y, Wu D, Trynka G, Raj T, Terao C, Ikari K, et al. Genetics of rheumatoid arthritis contributes to biology and drug discovery. Nature. 2014;506(7488):376–81. Epub 2014/01/07. 10.1038/nature12873
    1. Senhaji N, Serrano A, Badre W, Serbati N, Karkouri M, Zaid Y, et al. Association of inflammatory cytokine gene polymorphisms with inflammatory bowel disease in a Moroccan cohort. Genes Immun. 2015. Epub 2015/12/04.
    1. Bank S, Skytt Andersen P, Burisch J, Pedersen N, Roug S, Galsgaard J, et al. Polymorphisms in the inflammatory pathway genes TLR2, TLR4, TLR9, LY96, NFKBIA, NFKB1, TNFA, TNFRSF1A, IL6R, IL10, IL23R, PTPN22, and PPARG are associated with susceptibility of inflammatory bowel disease in a Danish cohort. PLoS One. 2014;9(6):e98815 Epub 2014/06/28. 10.1371/journal.pone.0098815
    1. Deloukas P, Kanoni S, Willenborg C, Farrall M, Assimes TL, Thompson JR, et al. Large-scale association analysis identifies new risk loci for coronary artery disease. Nature genetics. 2013;45(1):25–33. Epub 2012/12/04. 10.1038/ng.2480
    1. Garnache-Ottou F, Chaperot L, Biichle S, Ferrand C, Remy-Martin JP, Deconinck E, et al. Expression of the myeloid-associated marker CD33 is not an exclusive factor for leukemic plasmacytoid dendritic cells. Blood. 2005;105(3):1256–64. Epub 2004/09/25. 10.1182/blood-2004-06-2416
    1. Lambert JC, Ibrahim-Verbaas CA, Harold D, Naj AC, Sims R, Bellenguez C, et al. Meta-analysis of 74,046 individuals identifies 11 new susceptibility loci for Alzheimer's disease. Nature genetics. 2013;45(12):1452–8. Epub 2013/10/29. 10.1038/ng.2802
    1. Bertram L, Lange C, Mullin K, Parkinson M, Hsiao M, Hogan MF, et al. Genome-wide association analysis reveals putative Alzheimer's disease susceptibility loci in addition to APOE. American journal of human genetics. 2008;83(5):623–32. Epub 2008/11/04. 10.1016/j.ajhg.2008.10.008
    1. Bradshaw EM, Chibnik LB, Keenan BT, Ottoboni L, Raj T, Tang A, et al. CD33 Alzheimer's disease locus: altered monocyte function and amyloid biology. Nature neuroscience. 2013;16(7):848–50. Epub 2013/05/28. 10.1038/nn.3435
    1. Jiang T, Yu JT, Hu N, Tan MS, Zhu XC, Tan L. CD33 in Alzheimer's disease. Molecular neurobiology. 2014;49(1):529–35. Epub 2013/08/29. 10.1007/s12035-013-8536-1
    1. Hollingworth P, Harold D, Sims R, Gerrish A, Lambert JC, Carrasquillo MM, et al. Common variants at ABCA7, MS4A6A/MS4A4E, EPHA1, CD33 and CD2AP are associated with Alzheimer's disease. Nature genetics. 2011;43(5):429–35. Epub 2011/04/05. 10.1038/ng.803
    1. Naj AC, Jun G, Beecham GW, Wang LS, Vardarajan BN, Buros J, et al. Common variants at MS4A4/MS4A6E, CD2AP, CD33 and EPHA1 are associated with late-onset Alzheimer's disease. Nature genetics. 2011;43(5):436–41. Epub 2011/04/05. 10.1038/ng.801
    1. Liu JZ, van Sommeren S, Huang H, Ng SC, Alberts R, Takahashi A, et al. Association analyses identify 38 susceptibility loci for inflammatory bowel disease and highlight shared genetic risk across populations. Nature genetics. 2015;47(9):979–86. Epub 2015/07/21. 10.1038/ng.3359
    1. Greenawalt DM, Dobrin R, Chudin E, Hatoum IJ, Suver C, Beaulaurier J, et al. A survey of the genetics of stomach, liver, and adipose gene expression from a morbidly obese cohort. Genome research. 2011;21(7):1008–16. Epub 2011/05/24. 10.1101/gr.112821.110
    1. Welter D, MacArthur J, Morales J, Burdett T, Hall P, Junkins H, et al. The NHGRI GWAS Catalog, a curated resource of SNP-trait associations. Nucleic acids research. 2014;42(Database issue):D1001–6. Epub 2013/12/10. 10.1093/nar/gkt1229
    1. Marquez A, Cenit MC, Nunez C, Mendoza JL, Taxonera C, Diaz-Rubio M, et al. Effect of BSN-MST1 locus on inflammatory bowel disease and multiple sclerosis susceptibility. Genes Immun. 2009;10(7):631–5. Epub 2009/08/07. 10.1038/gene.2009.56
    1. Rossin EJ, Lage K, Raychaudhuri S, Xavier RJ, Tatar D, Benita Y, et al. Proteins encoded in genomic regions associated with immune-mediated disease physically interact and suggest underlying biology. PLoS genetics. 2011;7(1):e1001273 10.1371/journal.pgen.1001273
    1. Häuser F, Deyle C, Berard D, Neukirch C, Glowacki C, Bickmann J, et al. Macrophage-stimulating protein polymorphism rs3197999 is associated with a gain of function: implications for inflammatory bowel disease. Genes and immunity. 2012;13(4):321–7. 10.1038/gene.2011.88
    1. Hauser F, Rossmann H, Laubert-Reh D, Wild PS, Zeller T, Muller C, et al. Inflammatory bowel disease (IBD) locus 12: is glutathione peroxidase-1 (GPX1) the relevant gene? Genes Immun. 2015;16(8):571–5. Epub 2015/09/12. 10.1038/gene.2015.35
    1. Sandborn WJ, Feagan BG, Marano C, Zhang H, Strauss R, Johanns J, et al. Subcutaneous golimumab induces clinical response and remission in patients with moderate-to-severe ulcerative colitis. Gastroenterology. 2014;146(1):85–95; quiz e14-5. Epub 2013/06/06. 10.1053/j.gastro.2013.05.048
    1. Sandborn WJ, Gasink C, Gao LL, Blank MA, Johanns J, Guzzo C, et al. Ustekinumab induction and maintenance therapy in refractory Crohn's disease. The New England journal of medicine. 2012;367(16):1519–28. Epub 2012/10/19. 10.1056/NEJMoa1203572
    1. Gold L, Ayers D, Bertino J, Bock C, Bock A, Brody EN, et al. Aptamer-based multiplexed proteomic technology for biomarker discovery. PloS one. 2010;5(12):e15004 10.1371/journal.pone.0015004
    1. Howie B, Fuchsberger C, Stephens M, Marchini J, Abecasis GR. Fast and accurate genotype imputation in genome-wide association studies through pre-phasing. Nature genetics. 2012;44(8):955–9. Epub 2012/07/24. 10.1038/ng.2354
    1. Schadt EE, Molony C, Chudin E, Hao K, Yang X, Lum PY, et al. Mapping the genetic architecture of gene expression in human liver. PLoS biology. 2008;6(5):e107 Epub 2008/05/09. 10.1371/journal.pbio.0060107
    1. Hao K, Schadt EE, Storey JD. Calibrating the performance of SNP arrays for whole-genome association studies. PLoS genetics. 2008;4(6):e1000109 10.1371/journal.pgen.1000109
    1. Hao K, Bossé Y, Nickle DC, Paré PD, Postma DS, Laviolette M, et al. Lung eQTLs to help reveal the molecular underpinnings of asthma. PLoS genetics. 2012;8(11):e1003029 10.1371/journal.pgen.1003029

Source: PubMed

3
Tilaa