Subphenotypes of inflammatory bowel disease are characterized by specific serum protein profiles

Erik Andersson, Daniel Bergemalm, Robert Kruse, Gunter Neumann, Mauro D'Amato, Dirk Repsilber, Jonas Halfvarson, Erik Andersson, Daniel Bergemalm, Robert Kruse, Gunter Neumann, Mauro D'Amato, Dirk Repsilber, Jonas Halfvarson

Abstract

Objective: Genetic and immunological data indicate that inflammatory bowel disease (IBD) are characterized by specific inflammatory protein profiles. However, the serum proteome of IBD is still to be defined. We aimed to characterize the inflammatory serum protein profiles of Crohn's disease (CD) and ulcerative colitis (UC), using the novel proximity extension assay.

Methods: A panel of 91 inflammatory proteins were quantified in a discovery cohort of CD (n = 54), UC patients (n = 54), and healthy controls (HCs; n = 54). We performed univariate analyses by t-test, with false discovery rate correction. A sparse partial least-squares (sPLS) approach was used to identify additional discriminative proteins. The results were validated in a replication cohort.

Results: By univariate analysis, 17 proteins were identified with significantly different abundances in CD and HCs, and 12 when comparing UC and HCs. Additionally, 64 and 45 discriminant candidate proteins, respectively, were identified with the multivariate approach. Correspondingly, significant cross-validation error rates of 0.12 and 0.19 were observed in the discovery cohort. Only FGF-19 was identified from univariate comparisons of CD and UC, but 37 additional discriminant candidates were identified using the multivariate approach. The observed cross-validation error rate for CD vs. UC remained significant when restricting the analyses to patients in clinical remission. Using univariate comparisons, 16 of 17 CD-associated proteins and 8 of 12 UC-associated proteins were validated in the replication cohort. The area under the curve for CD and UC was 0.96 and 0.92, respectively, when the sPLS model from the discovery cohort was applied to the replication cohort.

Conclusions: By using the novel PEA method and a panel of inflammatory proteins, we identified proteins with significantly different quantities in CD patients and UC patients compared to HCs. Our data highlight the potential of the serum IBD proteome as a source for identification of future diagnostic biomarkers.

Conflict of interest statement

Competing Interests: JH has received research grants from the Swedish Research Council, the Örebro University Hospital Research Foundation, the Swedish Foundation for Strategic Research, and speaker's and/or consultancy fees from Abbvie, Cellgene, Hospira, Janssen, Medivir, MSD, Pfizer, Vifor Pharma, Takeda and Tillotts Pharma. DB has received grants from Bengt Ihre research foundation and grants from Region Örebro county, during the conduct of the study. EA, RK, GN, DR, MD have no personal conflicts of interests to declare. This does not alter our adherence to PLOS ONE policies on sharing data and materials.

Figures

Fig 1. Venn diagram of significantly altered…
Fig 1. Venn diagram of significantly altered proteins (q 20%) from comparison of UC patients vs. healthy controls (HCs) and CD patients vs. HCs in the discovery cohort.
Seven markers were common between the analyses.
Fig 2. sPLS plot showing the distribution…
Fig 2. sPLS plot showing the distribution of all samples from CD patients, UC patients, and healthy controls (HCs) in the discovery cohort, based on the 20 most important proteins from the multivariate analysis.
Fig 3. sPLS plot showing the distribution…
Fig 3. sPLS plot showing the distribution of patients with ileal CD, colonic CD, and UC in the discovery cohort, based on the 20 most important proteins from the multivariate analysis.
Fig 4. Validated proteins.
Fig 4. Validated proteins.
Significant biomarkers (q 20%) from the discovery cohort that passed validation in the replication cohort shown with fold changes and 95% confidence intervals. Actual fold changes from the replication cohort are presented with a red dot in the graph. A: Proteins validated in CD versus HC samples. B: Validated proteins in UC versus HC samples.
Fig 5. Venn diagram of validated proteins…
Fig 5. Venn diagram of validated proteins from comparison of UC patients vs. healthy controls (HCs) and CD patients vs. HCs in the replication cohort.
Ten proteins were unique to CD and two were unique to UC.
Fig 6. Receiver operator characteristics curves (ROC)…
Fig 6. Receiver operator characteristics curves (ROC) of the sparse partial least squares models applied on the replication cohort, with area under the curves (AUCs).
L2, colonic Crohn’s disease; L1/L3, ileal/ileocolonic Crohn’s disease; CD in remission, Crohn’s disease patients in clinical remission; UC in remission; ulcerative colitis patients in clinical remission.

References

    1. de Souza HS, Fiocchi C. Immunopathogenesis of IBD: current state of the art. Nature reviews Gastroenterology & hepatology. 2016;13(1):13–27. Epub 2015/12/03. doi: .
    1. Ek WE, D'Amato M, Halfvarson J. The history of genetics in inflammatory bowel disease. Annals of gastroenterology: quarterly publication of the Hellenic Society of Gastroenterology. 2014;27(4):294–303. Epub 2014/10/22.
    1. Ungaro R, Mehandru S, Allen PB, Peyrin-Biroulet L, Colombel J-F. Ulcerative colitis. The Lancet. .
    1. Torres J, Mehandru S, Colombel J-F, Peyrin-Biroulet L. Crohn's disease. The Lancet. .
    1. Subramanian S, Ekbom A, Rhodes JM. Recent advances in clinical practice: a systematic review of isolated colonic Crohn's disease: the third IBD? Gut. 2017;66(2):362–81. Epub 2016/11/02. doi: .
    1. Cleynen I, Boucher G, Jostins L, Schumm LP, Zeissig S, Ahmad T, et al. Inherited determinants of Crohn's disease and ulcerative colitis phenotypes: a genetic association study. Lancet (London, England). 2016;387(10014):156–67. Epub 2015/10/23. doi:
    1. Ray S, Reddy PJ, Jain R, Gollapalli K, Moiyadi A, Srivastava S. Proteomic technologies for the identification of disease biomarkers in serum: advances and challenges ahead. Proteomics. 2011;11(11):2139–61. Epub 2011/05/07. doi: .
    1. Assarsson E, Lundberg M, Holmquist G, Bjorkesten J, Thorsen SB, Ekman D, et al. Homogenous 96-plex PEA immunoassay exhibiting high sensitivity, specificity, and excellent scalability. PloS one. 2014;9(4):e95192 Epub 2014/04/24. doi:
    1. Fransen K, Franzen P, Magnuson A, Elmabsout AA, Nyhlin N, Wickbom A, et al. Polymorphism in the retinoic acid metabolizing enzyme CYP26B1 and the development of Crohn's Disease. PloS one. 2013;8(8):e72739 Epub 2013/08/27. doi:
    1. Lennard-Jones JE. Classification of inflammatory bowel disease. Scandinavian journal of gastroenterology Supplement. 1989;170:2–6; discussion 16–9. Epub 1989/01/01. .
    1. Silverberg MS, Satsangi J, Ahmad T, Arnott ID, Bernstein CN, Brant SR, et al. Toward an integrated clinical, molecular and serological classification of inflammatory bowel disease: report of a Working Party of the 2005 Montreal World Congress of Gastroenterology. Canadian journal of gastroenterology = Journal canadien de gastroenterologie. 2005;19 Suppl A:5a–36a. Epub 2005/09/10. .
    1. Harvey RF, Bradshaw JM. A simple index of Crohn's-disease activity. Lancet (London, England). 1980;1(8167):514 Epub 1980/03/08. .
    1. Lewis JD, Chuai S, Nessel L, Lichtenstein GR, Aberra FN, Ellenberg JH. Use of the noninvasive components of the Mayo score to assess clinical response in ulcerative colitis. Inflamm Bowel Dis. 2008;14(12):1660–6. Epub 2008/07/16. doi:
    1. Lê Cao K-A, Boitard S, Besse P. Sparse PLS discriminant analysis: Biologically relevant feature selection and graphical displays for multiclass problems. BMC Bioinformatics. 2011;12(1):253-. doi:
    1. Eriksson L. Multi- and megavariate data analysis P. 1, Basic principles and applications. 2nd ed Umeå: Umetrics Academy; 2006.
    1. Wold S. PLS for multivariate linear modeling. Chemometric methods in molecular design. 1995;2:195.
    1. Mehmood T, Warringer J, Snipen L, Sæbø S, Faculty of S, Naturvetenskapliga f, et al. Improving stability and understandability of genotype-phenotype mapping in Saccharomyces using regularized variable selection in L-PLS regression. BMC Bioinformatics. 2012;13(1):327-. doi:
    1. Coburn LA, Horst SN, Chaturvedi R, Brown CT, Allaman MM, Scull BP, et al. High-throughput multi-analyte Luminex profiling implicates eotaxin-1 in ulcerative colitis. PloS one. 2013;8(12):e82300 Epub 2013/12/25. doi:
    1. Rodriguez-Peralvarez ML, Garcia-Sanchez V, Villar-Pastor CM, Gonzalez R, Iglesias-Flores E, Muntane J, et al. Role of serum cytokine profile in ulcerative colitis assessment. Inflamm Bowel Dis. 2012;18(10):1864–71. Epub 2012/01/13. doi: .
    1. Korolkova OY, Myers JN, Pellom ST, Wang L, M'Koma AE. Characterization of Serum Cytokine Profile in Predominantly Colonic Inflammatory Bowel Disease to Delineate Ulcerative and Crohn's Colitides. Clinical medicine insights Gastroenterology. 2015;8:29–44. Epub 2015/06/17. doi:
    1. Townsend P, Zhang Q, Shapiro J, Webb-Robertson BJ, Bramer L, Schepmoes AA, et al. Serum Proteome Profiles in Stricturing Crohn's Disease: A Pilot Study. Inflamm Bowel Dis. 2015;21(8):1935–41. Epub 2015/07/23. doi:
    1. Li N, Wang X, Zhang Y, Zhai J, Zhang T, Wei K. Comparative proteomics analysis of serum proteins in ulcerative colitis patients. Molecular biology reports. 2012;39(5):5659–67. Epub 2011/12/27. doi: .
    1. Meuwis MA, Fillet M, Geurts P, de Seny D, Lutteri L, Chapelle JP, et al. Biomarker discovery for inflammatory bowel disease, using proteomic serum profiling. Biochemical pharmacology. 2007;73(9):1422–33. Epub 2007/01/30. doi: .
    1. Wasinger VC, Yau Y, Duo X, Zeng M, Campbell B, Shin S, et al. Low Mass Blood Peptides Discriminative of Inflammatory Bowel Disease (IBD) Severity: A Quantitative Proteomic Perspective. Molecular & cellular proteomics: MCP. 2016;15(1):256–65. Epub 2015/11/05. doi:
    1. Lacher M, Kappler R, Berkholz S, Baurecht H, von Schweinitz D, Koletzko S. Association of a CXCL9 polymorphism with pediatric Crohn's disease. Biochemical and biophysical research communications. 2007;363(3):701–7. Epub 2007/09/26. doi: .
    1. Hedl M, Abraham C. A TPL2 (MAP3K8) disease-risk polymorphism increases TPL2 expression thereby leading to increased pattern recognition receptor-initiated caspase-1 and caspase-8 activation, signalling and cytokine secretion. Gut. 2015. Epub 2015/07/29. doi: .
    1. Singh UP, Singh NP, Murphy EA, Price RL, Fayad R, Nagarkatti M, et al. Chemokine and cytokine levels in inflammatory bowel disease patients. Cytokine. 2016;77:44–9. Epub 2015/11/02. doi:
    1. Foell D, Kucharzik T, Kraft M, Vogl T, Sorg C, Domschke W, et al. Neutrophil derived human S100A12 (EN-RAGE) is strongly expressed during chronic active inflammatory bowel disease. Gut. 2003;52(6):847–53. Epub 2003/05/13.
    1. Kaiser T, Langhorst J, Wittkowski H, Becker K, Friedrich AW, Rueffer A, et al. Faecal S100A12 as a non-invasive marker distinguishing inflammatory bowel disease from irritable bowel syndrome. Gut. 2007;56(12):1706–13. Epub 2007/08/07. doi:
    1. Dabritz J, Langhorst J, Lugering A, Heidemann J, Mohr M, Wittkowski H, et al. Improving relapse prediction in inflammatory bowel disease by neutrophil-derived S100A12. Inflamm Bowel Dis. 2013;19(6):1130–8. Epub 2013/02/05. doi: .
    1. Moschen AR, Kaser A, Enrich B, Ludwiczek O, Gabriel M, Obrist P, et al. The RANKL/OPG system is activated in inflammatory bowel disease and relates to the state of bone loss. Gut. 2005;54(4):479–87. Epub 2005/03/09. doi:
    1. Stanislawowski M, Wisniewski P, Guzek M, Wierzbicki PM, Adrych K, Smoczynski M, et al. Influence of receptor activator of nuclear factor kappa B ligand, osteoprotegerin and interleukin-33 on bone metabolism in patients with long-standing ulcerative colitis. Journal of Crohn's & colitis. 2014;8(8):802–10. Epub 2014/01/21. doi: .
    1. Beigel F, Friedrich M, Probst C, Sotlar K, Goke B, Diegelmann J, et al. Oncostatin M mediates STAT3-dependent intestinal epithelial restitution via increased cell proliferation, decreased apoptosis and upregulation of SERPIN family members. PloS one. 2014;9(4):e93498 Epub 2014/04/09. doi:
    1. Jungbeck M, Daller B, Federhofer J, Wege AK, Wimmer N, Mannel DN, et al. Neutralization of LIGHT ameliorates acute dextran sodium sulphate-induced intestinal inflammation. Immunology. 2009;128(3):451–8. Epub 2010/01/14. doi:
    1. Hormi K, Cadiot G, Kermorgant S, Dessirier V, Le Romancer M, Lewin MJ, et al. Transforming growth factor-alpha and epidermal growth factor receptor in colonic mucosa in active and inactive inflammatory bowel disease. Growth factors (Chur, Switzerland). 2000;18(2):79–91. Epub 2000/10/06. .
    1. Grip O, Malm J, Veress B, Bjartell A, Lindgren S, Egesten A. Increased presence of cells containing transforming growth factor alpha (TGF-alpha) in ulcerative colitis, both during active inflammation and in remission. European journal of gastroenterology & hepatology. 2000;12(7):761–6. Epub 2000/08/10. .
    1. Kurosu H, Choi M, Ogawa Y, Dickson AS, Goetz R, Eliseenkova AV, et al. Tissue-specific expression of betaKlotho and fibroblast growth factor (FGF) receptor isoforms determines metabolic activity of FGF19 and FGF21. The Journal of biological chemistry. 2007;282(37):26687–95. Epub 2007/07/12. doi:
    1. Nolan JD, Johnston IM, Pattni SS, Dew T, Orchard TR, Walters JR. Diarrhea in Crohn's disease: investigating the role of the ileal hormone fibroblast growth factor 19. Journal of Crohn's & colitis. 2015;9(2):125–31. Epub 2014/12/18. doi: .
    1. Yang D, Chen Q, Hoover DM, Staley P, Tucker KD, Lubkowski J, et al. Many chemokines including CCL20/MIP-3alpha display antimicrobial activity. Journal of leukocyte biology. 2003;74(3):448–55. Epub 2003/09/02. .
    1. Lazarus NH, Kunkel EJ, Johnston B, Wilson E, Youngman KR, Butcher EC. A common mucosal chemokine (mucosae-associated epithelial chemokine/CCL28) selectively attracts IgA plasmablasts. Journal of immunology (Baltimore, Md: 1950). 2003;170(7):3799–805. Epub 2003/03/21. .
    1. Falvey JD, Hoskin T, Meijer B, Ashcroft A, Walmsley R, Day AS, et al. Disease activity assessment in IBD: clinical indices and biomarkers fail to predict endoscopic remission. Inflamm Bowel Dis. 2015;21(4):824–31. Epub 2015/03/05. doi: .

Source: PubMed

3
Se inscrever