Skip to content
Licensed Unlicensed Requires Authentication Published by De Gruyter December 13, 2019

Complexity, cost, and content – three important factors for translation of clinical protein mass spectrometry tests, and the case for apolipoprotein C-III proteoform testing

  • Dobrin Nedelkov EMAIL logo and Yueming Hu

Abstract

Complexity, cost, and content are three important factors that can impede translation of clinical protein mass spectrometry (MS) tests at a larger scale. Complexity stems from the many components/steps involved in bottom-up protein MS workflows, making them significantly more complicated than enzymatic immunoassays (EIA) that currently dominate clinical testing. This complexity inevitably leads to increased costs, which is detrimental in the price-competitive clinical marketplace. To successfully compete, new clinical protein MS tests need to offer something new and unique that EIAs cannot – a new content of proteoform detection. The preferred method for proteoform profiling is intact protein MS analysis, in which all proteins are measured as intact species thus allowing discovery of new proteoforms. To illustrate the importance of intact proteoform testing with MS and its potential clinical implications, we discuss here recent findings from multiple studies on the distribution of apolipoprotein C-III proteoforms and their correlations with key clinical measures of dyslipidemia. Such studies are only made possible with assays that are low in cost, avoid unnecessary complexity, and are unique in providing the content of proteoforms.

  1. Author contributions: All the authors have accepted responsibility for the entire content of this submitted manuscript and approved submission.

  2. Research funding: None declared.

  3. Employment or leadership: None declared.

  4. Honorarium: None declared.

  5. Competing interests: The funding organization(s) played no role in the study design; in the collection, analysis, and interpretation of data; in the writing of the report; or in the decision to submit the report for publication.

References

1. Nedelkov D. Mass spectrometry protein tests: ready for prime time (or not). Expert Rev Proteomics 2017;14:1–7.10.1080/14789450.2017.1256777Search in Google Scholar PubMed

2. Ombrone D, Giocaliere E, Forni G, Malvagia S, la Marca G. Expanded newborn screening by mass spectrometry: new tests, future perspectives. Mass Spectrom Rev 2016;35:71–84.10.1002/mas.21463Search in Google Scholar PubMed

3. Van Wijk XM, Goodnough R, Colby JM. Mass spectrometry in emergency toxicology: current state and future applications. Crit Rev Clin Lab Sci 2019;56:225–38.10.1080/10408363.2019.1585415Search in Google Scholar PubMed

4. Lequin RM. Enzyme immunoassay (EIA)/enzyme-linked immunosorbent assay (ELISA). Clin Chem 2005;51:2415–8.10.1373/clinchem.2005.051532Search in Google Scholar PubMed

5. Marquette CA, Corgier BP, Blum LJ. Recent advances in multiplex immunoassays. Bioanalysis 2012;4:927–36.10.4155/bio.12.56Search in Google Scholar PubMed

6. Percy AJ, Byrns S, Pennington SR, Holmes DT, Anderson NL, Agreste TM, et al. Clinical translation of MS-based, quantitative plasma proteomics: status, challenges, requirements, and potential. Expert Rev Proteomics 2016;13:673–84.10.1080/14789450.2016.1205950Search in Google Scholar PubMed

7. Manes NP, Nita-Lazar A. Application of targeted mass spectrometry in bottom-up proteomics for systems biology research. J Proteomics 2018;189:75–90.10.1016/j.jprot.2018.02.008Search in Google Scholar PubMed PubMed Central

8. van de Merbel NC. Protein quantification by LC-MS: a decade of progress through the pages of. Bioanalysis 2019;11:629–44.10.4155/bio-2019-0032Search in Google Scholar PubMed

9. Hüttenhain R, Choi M, Martin de la Fuente L, Oehl K, Chang CV, Zimmermann AK, et al. A targeted mass spectrometry strategy for developing proteomic biomarkers: a case study of epithelial ovarian cancer. Mol Cell Proteomics 2019;18:1836–50.10.1074/mcp.RA118.001221Search in Google Scholar PubMed PubMed Central

10. Shuford CM, Walters JJ, Holland PM, Sreenivasan U, Askari N, Ray K, et al. Absolute protein quantification by mass spectrometry: not as simple as advertised. Anal Chem 2017;89:7406–15.10.1021/acs.analchem.7b00858Search in Google Scholar PubMed

11. Jeffs JW, Ferdosi S, Yassine HN, Borges CR. Ex vivo instability of glycated albumin: a role for autoxidative glycation. Arch Biochem Biophys 2017;629:36–42.10.1016/j.abb.2017.07.004Search in Google Scholar

12. Hattan SJ, Parker KC, Vestal ML, Yang JY, Herold DA, Duncan MW. Analysis and quantitation of glycated hemoglobin by matrix assisted laser desorption/ionization time of flight mass spectrometry. J Am Soc Mass Spectrom 2016;27:532–41.10.1007/s13361-015-1316-6Search in Google Scholar

13. Smith LM, Kelleher NL, Proteomics CF. Proteoform: a single term describing protein complexity. Nat Methods 2013;10:186–7.10.1038/nmeth.2369Search in Google Scholar

14. Roy B, Haupt LM, Griffiths LR. Review: alternative splicing (AS) of genes as an approach for generating protein complexity. Curr Genomics 2013;14:182–94.10.2174/1389202911314030004Search in Google Scholar

15. Wu JR, Zeng R. Molecular basis for population variation: from SNPs to SAPs. FEBS Lett 2012;586:2841–5.10.1016/j.febslet.2012.07.036Search in Google Scholar

16. Farley AR, Link AJ. Identification and quantification of protein posttranslational modifications. Methods Enzymol 2009;463:725–63.10.1016/S0076-6879(09)63040-8Search in Google Scholar

17. Nedelkov D. Human proteoforms as new targets for clinical mass spectrometry protein tests. Expert Rev Proteomics 2017;14:691–9.10.1080/14789450.2017.1362337Search in Google Scholar PubMed

18. Bystrom C, Sheng S, Zhang K, Caulfield M, Clarke NJ, Reitz R. Clinical utility of insulin-like growth factor 1 and 2; determination by high resolution mass spectrometry. PLoS One 2012;7:e43457.10.1371/journal.pone.0043457Search in Google Scholar PubMed PubMed Central

19. Chen Z, Caulfield MP, McPhaul MJ, Reitz RE, Taylor SW, Clarke NJ. Quantitative insulin analysis using liquid chromatography-tandem mass spectrometry in a high-throughput clinical laboratory. Clin Chem 2013;59:1349–56.10.1373/clinchem.2012.199794Search in Google Scholar PubMed

20. Hoofnagle AN, Roth MY. Clinical review: improving the measurement of serum thyroglobulin with mass spectrometry. J Clin Endocrinol Metab 2013;98:1343–52.10.1210/jc.2012-4172Search in Google Scholar PubMed PubMed Central

21. Lacey JM, Bergen HR, Magera MJ, Naylor S, O’Brien JF. Rapid determination of transferrin isoforms by immunoaffinity liquid chromatography and electrospray mass spectrometry. Clin Chem 2001;47:513–8.10.1093/clinchem/47.3.513Search in Google Scholar

22. Bergen HR, Zeldenrust SR, Butz ML, Snow DS, Dyck PJ, Klein CJ, et al. Identification of transthyretin variants by sequential proteomic and genomic analysis. Clin Chem 2004;50:1544–52.10.1373/clinchem.2004.033266Search in Google Scholar PubMed

23. Genzen JR, Mohlman JS, Lynch JL, Squires MW, Weiss RL. Laboratory-developed tests: a legislative and regulatory review. Clin Chem 2017;63:1575–84.10.1373/clinchem.2017.275164Search in Google Scholar PubMed

24. Lenters-Westra E, Schindhelm RK, Bilo HJ, Slingerland RJ. Haemoglobin A1c: historical overview and current concepts. Diabetes Res Clin Pract 2013;99:75–84.10.1016/j.diabres.2012.10.007Search in Google Scholar PubMed

25. Xu A, Wang Y, Li J, Liu G, Li X, Chen W, et al. Evaluation of MALDI-TOF MS for the measurement of glycated hemoglobin. Clin Chim Acta 2019;498:154–60.10.1016/j.cca.2019.08.025Search in Google Scholar PubMed

26. Tubbs KA, Nedelkov D, Nelson RW. Detection and quantification of beta-2-microglobulin using mass spectrometric immunoassay. Anal Biochem 2001;289:26–35.10.1006/abio.2000.4921Search in Google Scholar PubMed

27. Trenchevska O, Nelson R, Nedelkov D. Mass spectrometric immunoassays in characterization of clinically significant proteoforms. Proteomes 2016;4:13.10.3390/proteomes4010013Search in Google Scholar PubMed PubMed Central

28. Nedelkov D, Kiernan UA, Niederkofler EE, Tubbs KA, Nelson RW. Investigating diversity in human plasma proteins. Proc Natl Acad Sci U S A 2005;102:10852–7.10.1073/pnas.0500426102Search in Google Scholar PubMed PubMed Central

29. Wyler von Ballmoos MC, Haring B, Sacks FM. The risk of cardiovascular events with increased apolipoprotein CIII: a systematic review and meta-analysis. J Clin Lipidol 2015;9:498–510.10.1016/j.jacl.2015.05.002Search in Google Scholar PubMed

30. Norata GD, Tsimikas S, Pirillo A, Catapano AL. Apolipoprotein C-III: from pathophysiology to pharmacology. Trends Pharmacol Sci 2015;36:675–87.10.1016/j.tips.2015.07.001Search in Google Scholar PubMed

31. Ramms B, Gordts PL. Apolipoprotein C-III in triglyceride-rich lipoprotein metabolism. Curr Opin Lipidol 2018;29:171–9.10.1097/MOL.0000000000000502Search in Google Scholar

32. Crosby J, Peloso GM, Auer PL, Crosslin DR, Stitziel NO, Lange LA, et al. Loss-of-function mutations in APOC3, triglycerides, and coronary disease. N Engl J Med 2014;371:22–31.10.1056/NEJMoa1307095Search in Google Scholar

33. Jørgensen AB, Frikke-Schmidt R, Nordestgaard BG, Tybjærg-Hansen A. Loss-of-function mutations in APOC3 and risk of ischemic vascular disease. N Engl J Med 2014;371:32–41.10.1056/NEJMoa1308027Search in Google Scholar

34. Graham MJ, Lee RG, Bell TA, Fu W, Mullick AE, Alexander VJ, et al. Antisense oligonucleotide inhibition of apolipoprotein C-III reduces plasma triglycerides in rodents, nonhuman primates, and humans. Circ Res 2013;112:1479–90.10.1161/CIRCRESAHA.111.300367Search in Google Scholar

35. Schmitz J, Gouni-Berthold I. APOC-III antisense oligonucleotides: a new option for the treatment of hypertriglyceridemia. Curr Med Chem 2018;25:1567–76.10.2174/0929867324666170609081612Search in Google Scholar

36. Juntti-Berggren L, Berggren PO. Apolipoprotein CIII is a new player in diabetes. Curr Opin Lipidol 2017;28:27–31.10.1097/MOL.0000000000000372Search in Google Scholar

37. Zhang J, Rocha NA, McCullough PA. Contribution of ApoCIII to diabetic dyslipidemia and treatment with volanesorsen. Rev Cardiovasc Med 2018;19:13–9.10.31083/j.rcm.2018.01.890Search in Google Scholar

38. Bondarenko PV, Cockrill SL, Watkins LK, Cruzado ID, Macfarlane RD. Mass spectral study of polymorphism of the apolipoproteins of very low density lipoprotein. J Lipid Res 1999;40:543–55.10.1016/S0022-2275(20)32459-7Search in Google Scholar

39. Wada Y, Kadoya M, Okamoto N. Mass spectrometry of apolipoprotein C-III, a simple analytical method for mucin- type O-glycosylation and its application to an autosomal recessive cutis laxa type-2 (ARCL2) patient. Glycobiology 2012;22:1140–4.10.1093/glycob/cws086Search in Google Scholar PubMed

40. Jian W, Edom RW, Wang D, Weng N, Zhang SW. Relative quantitation of glycoisoforms of intact apolipoprotein C3 in human plasma by liquid chromatography-high-resolution mass spectrometry. Anal Chem 2013;85:2867–74.10.1021/ac3034757Search in Google Scholar PubMed

41. Nicolardi S, van der Burgt YE, Dragan I, Hensbergen PJ, Deelder AM. Identification of new apolipoprotein-CIII glycoforms with ultrahigh resolution MALDI-FTICR mass spectrometry of human sera. J Proteome Res 2013;12:2260–8.10.1021/pr400136pSearch in Google Scholar PubMed

42. Trenchevska O, Schaab MR, Nelson RW, Nedelkov D. Development of multiplex mass spectrometric immunoassay for detection and quantification of apolipoproteins C-I, C-II, C-III and their proteoforms. Methods 2015;81:86–92.10.1016/j.ymeth.2015.02.020Search in Google Scholar PubMed PubMed Central

43. Yen-Nicolaÿ S, Boursier C, Rio M, Lefeber DJ, Pilon A, Seta N, et al. MALDI-TOF MS applied to apoC-III glycoforms of patients with congenital disorders affecting O-glycosylation. Comparison with two-dimensional electrophoresis. Proteomics Clin Appl 2015;9:787–93.10.1002/prca.201400187Search in Google Scholar PubMed

44. Ceglarek U, Dittrich J, Becker S, Baumann F, Kortz L, Thiery J. Quantification of seven apolipoproteins in human plasma by proteotypic peptides using fast LC-MS/MS. Proteomics Clin Appl 2013;7:794–801.10.1002/prca.201300034Search in Google Scholar PubMed

45. van den Broek I, Romijn FP, Nouta J, van der Laarse A, Drijfhout JW, Smit NP, et al. Automated multiplex LC-MS/MS assay for quantifying serum apolipoproteins A-I, B, C-I, C-II, C-III, and E with qualitative apolipoprotein E phenotyping. Clin Chem 2016;62:188–97.10.1373/clinchem.2015.246702Search in Google Scholar PubMed

46. Toth CA, Kuklenyik Z, Jones JI, Parks BA, Gardner MS, Schieltz DM, et al. On-column trypsin digestion coupled with LC-MS/MS for quantification of apolipoproteins. J Proteomics 2017;150:258–67.10.1016/j.jprot.2016.09.011Search in Google Scholar PubMed

47. Dittrich J, Adam M, Maas H, Hecht M, Reinicke M, Ruhaak LR, et al. Targeted on-line SPE-LC-MS/MS assay for the quantitation of 12 apolipoproteins from human blood. Proteomics 2018;18.10.1002/pmic.201700279Search in Google Scholar PubMed

48. Ruhaak LR, Smit NP, Romijn FP, Pieterse MM, van der Laarse A, van der Burgt YE, et al. Robust and accurate 2-year performance of a quantitative mass spectrometry-based apolipoprotein test in a clinical chemistry laboratory. Clin Chem 2018;64:747–9.10.1373/clinchem.2017.285098Search in Google Scholar PubMed

49. Nelson RW, Borges CR. Mass spectrometric immunoassay revisited. J Am Soc Mass Spectrom 2011;22:960–8.10.1007/s13361-011-0094-zSearch in Google Scholar PubMed PubMed Central

50. Yassine HN, Trenchevska O, Ramrakhiani A, Parekh A, Koska J, Walker RW, et al. The association of human apolipoprotein C-III sialylation proteoforms with plasma triglycerides. PLoS One 2015;10:e0144138.10.1371/journal.pone.0144138Search in Google Scholar PubMed PubMed Central

51. Koska J, Yassine H, Trenchevska O, Sinari S, Schwenke DC, Yen FT, et al. Disialylated apolipoprotein C-III proteoform is associated with improved lipids in prediabetes and type 2 diabetes. J Lipid Res 2016;57:894–905.10.1194/jlr.P064816Search in Google Scholar PubMed PubMed Central

52. Mendoza S, Trenchevska O, King SM, Nelson RW, Nedelkov D, Krauss RM, et al. Changes in low-density lipoprotein size phenotypes associate with changes in apolipoprotein C-III glycoforms after dietary interventions. J Clin Lipidol 2017;11:224–33.e2.10.1016/j.jacl.2016.12.009Search in Google Scholar PubMed PubMed Central

53. Kegulian NC, Ramms B, Horton S, Trenchevska O, Nedelkov D, Graham MJ, et al. ApoC-III glycoforms are differentially cleared by hepatic TRL (triglyceride-rich lipoprotein) receptors. Arterioscler Thromb Vasc Biol 2019;39:2145–56.10.1161/ATVBAHA.119.312723Search in Google Scholar PubMed PubMed Central

54. Cheng K, Chui H, Domish L, Hernandez D, Wang G. Recent development of mass spectrometry and proteomics applications in identification and typing of bacteria. Proteomics Clin Appl 2016;10:346–57.10.1002/prca.201500086Search in Google Scholar PubMed PubMed Central

55. Duncan MW, Nedelkov D, Walsh R, Hattan SJ. Applications of MALDI mass spectrometry in clinical chemistry. Clin Chem 2016;62:134–43.10.1373/clinchem.2015.239491Search in Google Scholar PubMed

Received: 2019-09-19
Accepted: 2019-11-15
Published Online: 2019-12-13
Published in Print: 2020-06-25

©2019 Walter de Gruyter GmbH, Berlin/Boston

Downloaded on 29.3.2024 from https://www.degruyter.com/document/doi/10.1515/cclm-2019-0977/html
Scroll to top button