Assessment of metabolomic and proteomic biomarkers in detection and prognosis of progression of renal function in chronic kidney disease

Esther Nkuipou-Kenfack, Flore Duranton, Nathalie Gayrard, Àngel Argilés, Ulrika Lundin, Klaus M Weinberger, Mohammed Dakna, Christian Delles, William Mullen, Holger Husi, Julie Klein, Thomas Koeck, Petra Zürbig, Harald Mischak, Esther Nkuipou-Kenfack, Flore Duranton, Nathalie Gayrard, Àngel Argilés, Ulrika Lundin, Klaus M Weinberger, Mohammed Dakna, Christian Delles, William Mullen, Holger Husi, Julie Klein, Thomas Koeck, Petra Zürbig, Harald Mischak

Abstract

Chronic kidney disease (CKD) is part of a number of systemic and renal diseases and may reach epidemic proportions over the next decade. Efforts have been made to improve diagnosis and management of CKD. We hypothesised that combining metabolomic and proteomic approaches could generate a more systemic and complete view of the disease mechanisms. To test this approach, we examined samples from a cohort of 49 patients representing different stages of CKD. Urine samples were analysed for proteomic changes using capillary electrophoresis-mass spectrometry and urine and plasma samples for metabolomic changes using different mass spectrometry-based techniques. The training set included 20 CKD patients selected according to their estimated glomerular filtration rate (eGFR) at mild (59.9±16.5 mL/min/1.73 m2; n = 10) or advanced (8.9±4.5 mL/min/1.73 m2; n = 10) CKD and the remaining 29 patients left for the test set. We identified a panel of 76 statistically significant metabolites and peptides that correlated with CKD in the training set. We combined these biomarkers in different classifiers and then performed correlation analyses with eGFR at baseline and follow-up after 2.8±0.8 years in the test set. A solely plasma metabolite biomarker-based classifier significantly correlated with the loss of kidney function in the test set at baseline and follow-up (ρ = -0.8031; p<0.0001 and ρ = -0.6009; p = 0.0019, respectively). Similarly, a urinary metabolite biomarker-based classifier did reveal significant association to kidney function (ρ = -0.6557; p = 0.0001 and ρ = -0.6574; p = 0.0005). A classifier utilising 46 identified urinary peptide biomarkers performed statistically equivalent to the urinary and plasma metabolite classifier (ρ = -0.7752; p<0.0001 and ρ = -0.8400; p<0.0001). The combination of both urinary proteomic and urinary and plasma metabolic biomarkers did not improve the correlation with eGFR. In conclusion, we found excellent association of plasma and urinary metabolites and urinary peptides with kidney function, and disease progression, but no added value in combining the different biomarkers data.

Conflict of interest statement

Competing Interests: H. Mischak is the founder and co-owner of Mosaiques Diagnostics, who developed the CE-MS technology for clinical application. E. Nkuipou-Kenfack, M. Dakna, J. Klein, T. Koeck, and P. Zürbig are employees of Mosaiques Diagnostics. U. Lundin was an employee of Biocrates Life Sciences AG. K. Weinberger is a shareholder and consultant of Biocrates Life Sciences AG, who developed the metabolomics platform and kit products for research and diagnostic applications. These issues do not alter the authors' adherence to all the PLOS ONE policies on sharing data and materials.

Figures

Figure 1. Regulation of metabolites and peptides.
Figure 1. Regulation of metabolites and peptides.
The fold changes of metabolites and peptides “mild CKD” vs. “advanced CKD”. A. Plasma metabolites. B. Urinary metabolites. C. Urinary peptides. C19∶0: Nonadecanoic acid. SM C26∶1: Sphingomyelin with acyl residue sum C26∶1. PC aa C42∶4: Phosphatidylcholine with acyl-alkyl residue sum C42∶4. C14∶2: Tetradecadienoylcarnitine. cis-C20∶1w9: cis-11-Eicosenoic acid. PC aa C42∶4: Phosphatidylcholine with acyl-alkyl residue sum C42∶4. C17∶0: Heptadecanoic acid. PC aa C42∶5: Phosphatidylcholine with acyl-alkyl residue sum C42∶5. C4: Nonanoylcarnitine. C5: Isovalerylcarnitine. ADMA: Asymmetric dimethylarginine. Total DMA: Total dimethylarginine. C9: Nonanoylcarnitine. C4∶1: Butenoylcarnitine. C5-DC(C6-OH): Acylcarnitine. C14∶1-OH: 3-Hydroxytetradecenoylcarnitine. dH: Deoxyhexose. HNAc(S2): (N-acetylhexosamine)-disulfate. C3∶1: Propenoylcarnitine. C7-DC: Pimelylcarnitine. H2-dH2: Dihexose-dideoxyhexose. Asn: Asparagine. Leu: Leucine. H1: Hexose. Pro: Proline. Cit: Citrulline.
Figure 2. Correlation analysis of metabolomic and…
Figure 2. Correlation analysis of metabolomic and proteomic based classifier scores with baseline eGFR.
The correlation analysis is performed by using the support vector machine classification scores obtained for the test set with baseline. A. Classifier MetaboP (plasma metabolites) ρ = −0.8031 and p

Figure 3. Correlation analysis of metabolomic and…

Figure 3. Correlation analysis of metabolomic and proteomic based classifier scores with follow-up eGFR.

The…

Figure 3. Correlation analysis of metabolomic and proteomic based classifier scores with follow-up eGFR.
The correlation analysis is performed by using the support vector machine classification scores obtained for the test set with follow-up eGFR. A. Classifier MetaboP (plasma metabolites) ρ = −0.6009 and p = 0.0019. B. Classifier MetaboU (urinary metabolites) ρ = −0.6574 and p = 0.0005. C. Classifier Pept (urinary peptides) ρ = −0.8400 and p

Figure 4. Correlation analysis of a combined…

Figure 4. Correlation analysis of a combined proteomics and metabolomics based classifier with baseline or…

Figure 4. Correlation analysis of a combined proteomics and metabolomics based classifier with baseline or follow-up eGFR.
A. Classifier Pept_MetaboP (urinary peptides and plasma metabolites) with baseline eGFR ρ = −0.7833 and p
Similar articles
Cited by
References
    1. National Kidney Foundation (2002) K/DOQI clinical practice guidelines for chronic kidney disease: evaluation, classification, and stratification. Am J Kidney Dis 39: S1–S226. - PubMed
    1. Miller WG, Bruns DE, Hortin GL, Sandberg S, Aakre KM, et al. (2009) Current issues in measurement and reporting of urinary albumin excretion. Clin Chem 55: 24–38. - PubMed
    1. Allison SJ (2013) Chronic kidney disease: The effect of age on CKD outcomes. Nat Rev Nephrol 9: 3. - PubMed
    1. Silberring J, Ciborowski P (2010) Biomarker discovery and clinical proteomics. Trends Analyt Chem 29: 128. - PMC - PubMed
    1. Deininger SO, Ebert MP, Futterer A, Gerhard M, Rocken C (2008) MALDI imaging combined with hierarchical clustering as a new tool for the interpretation of complex human cancers. J Proteome Res 7: 5230–5236. - PubMed
Show all 60 references
Publication types
MeSH terms
Related information
Grant support
The project was partly funded by grant GA 31354 (CodeAge) FP7-PEOPLE-2011-ITN program, GA 251368 (Protoclin) from the FP7-PEOPLE-2009-IAPP program, through EU-MASCARA (HEALTH-2011-278249), SysKID (HEALTH–F2–2009–241544), and the Eurotransbio Program. AA and HM are members of the European Uraemic Toxin working group of the European Society of Artificial Organs (ESAO). The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.
[x]
Cite
Copy Download .nbib
Format: AMA APA MLA NLM

NCBI Literature Resources

MeSH PMC Bookshelf Disclaimer

The PubMed wordmark and PubMed logo are registered trademarks of the U.S. Department of Health and Human Services (HHS). Unauthorized use of these marks is strictly prohibited.

Follow NCBI
Figure 3. Correlation analysis of metabolomic and…
Figure 3. Correlation analysis of metabolomic and proteomic based classifier scores with follow-up eGFR.
The correlation analysis is performed by using the support vector machine classification scores obtained for the test set with follow-up eGFR. A. Classifier MetaboP (plasma metabolites) ρ = −0.6009 and p = 0.0019. B. Classifier MetaboU (urinary metabolites) ρ = −0.6574 and p = 0.0005. C. Classifier Pept (urinary peptides) ρ = −0.8400 and p

Figure 4. Correlation analysis of a combined…

Figure 4. Correlation analysis of a combined proteomics and metabolomics based classifier with baseline or…

Figure 4. Correlation analysis of a combined proteomics and metabolomics based classifier with baseline or follow-up eGFR.
A. Classifier Pept_MetaboP (urinary peptides and plasma metabolites) with baseline eGFR ρ = −0.7833 and p
Similar articles
Cited by
References
    1. National Kidney Foundation (2002) K/DOQI clinical practice guidelines for chronic kidney disease: evaluation, classification, and stratification. Am J Kidney Dis 39: S1–S226. - PubMed
    1. Miller WG, Bruns DE, Hortin GL, Sandberg S, Aakre KM, et al. (2009) Current issues in measurement and reporting of urinary albumin excretion. Clin Chem 55: 24–38. - PubMed
    1. Allison SJ (2013) Chronic kidney disease: The effect of age on CKD outcomes. Nat Rev Nephrol 9: 3. - PubMed
    1. Silberring J, Ciborowski P (2010) Biomarker discovery and clinical proteomics. Trends Analyt Chem 29: 128. - PMC - PubMed
    1. Deininger SO, Ebert MP, Futterer A, Gerhard M, Rocken C (2008) MALDI imaging combined with hierarchical clustering as a new tool for the interpretation of complex human cancers. J Proteome Res 7: 5230–5236. - PubMed
Show all 60 references
Publication types
MeSH terms
Related information
Grant support
The project was partly funded by grant GA 31354 (CodeAge) FP7-PEOPLE-2011-ITN program, GA 251368 (Protoclin) from the FP7-PEOPLE-2009-IAPP program, through EU-MASCARA (HEALTH-2011-278249), SysKID (HEALTH–F2–2009–241544), and the Eurotransbio Program. AA and HM are members of the European Uraemic Toxin working group of the European Society of Artificial Organs (ESAO). The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.
[x]
Cite
Copy Download .nbib
Format: AMA APA MLA NLM
Figure 4. Correlation analysis of a combined…
Figure 4. Correlation analysis of a combined proteomics and metabolomics based classifier with baseline or follow-up eGFR.
A. Classifier Pept_MetaboP (urinary peptides and plasma metabolites) with baseline eGFR ρ = −0.7833 and p

References

    1. National Kidney Foundation (2002) K/DOQI clinical practice guidelines for chronic kidney disease: evaluation, classification, and stratification. Am J Kidney Dis 39: S1–S226.
    1. Miller WG, Bruns DE, Hortin GL, Sandberg S, Aakre KM, et al. (2009) Current issues in measurement and reporting of urinary albumin excretion. Clin Chem 55: 24–38.
    1. Allison SJ (2013) Chronic kidney disease: The effect of age on CKD outcomes. Nat Rev Nephrol 9: 3.
    1. Silberring J, Ciborowski P (2010) Biomarker discovery and clinical proteomics. Trends Analyt Chem 29: 128.
    1. Deininger SO, Ebert MP, Futterer A, Gerhard M, Rocken C (2008) MALDI imaging combined with hierarchical clustering as a new tool for the interpretation of complex human cancers. J Proteome Res 7: 5230–5236.
    1. Rodriguez-Suarez E, Siwy J, Zurbig P, Mischak H (2013) Urine as a source for clinical proteome analysis: From discovery to clinical application. Biochim Biophys Acta.
    1. Good DM, Zürbig P, Argiles A, Bauer HW, Behrens G, et al. (2010) Naturally occurring human urinary peptides for use in diagnosis of chronic kidney disease. Mol Cell Proteomics 9: 2424–2437.
    1. Zürbig P, Jerums G, Hovind P, MacIsaac R, Mischak H, et al. (2012) Urinary Proteomics for Early Diagnosis in Diabetic Nephropathy. Diabetes 61: 3304–3313.
    1. Roscioni SS, de ZD, Hellemons ME, Mischak H, Zurbig P, et al. (2012) A urinary peptide biomarker set predicts worsening of albuminuria in type 2 diabetes mellitus. Diabetologia 56: 259–267.
    1. Goek ON, Prehn C, Sekula P, Romisch-Margl W, Doring A, et al. (2013) Metabolites associate with kidney function decline and incident chronic kidney disease in the general population. Nephrol Dial Transplant 28: 2131–2138.
    1. Rhee EP, Clish CB, Ghorbani A, Larson MG, Elmariah S, et al. (2013) A combined epidemiologic and metabolomic approach improves CKD prediction. J Am Soc Nephrol 24: 1330–1338.
    1. Posada-Ayala M, Zubiri I, Martin-Lorenzo M, Sanz-Maroto A, Molero D, et al. (2014) Identification of a urine metabolomic signature in patients with advanced-stage chronic kidney disease. Kidney Int 85: 103–111.
    1. Argiles A, Siwy J, Duranton F, Gayrard N, Dakna M, et al. (2013) CKD273, a New Proteomics Classifier Assessing CKD and Its Prognosis. PLoS One 8: e62837.
    1. Duranton F, Lundin U, Gayrard N, Mischak H, Aparicio M, et al... (2013) Plasma and Urinary Amino Acid Metabolomic Profiling in Patients with Different Levels of Kidney Function. Clin J Am Soc Nephrol.
    1. Levey AS, Coresh J, Greene T, Stevens LA, Zhang YL, et al. (2006) Using standardized serum creatinine values in the modification of diet in renal disease study equation for estimating glomerular filtration rate. Ann Intern Med 145: 247–254.
    1. Mischak H, Allmaier G, Apweiler R, Attwood T, Baumann M, et al. (2010) Recommendations for biomarker identification and qualification in clinical proteomics. Sci Transl Med 2: 46ps42.
    1. Mischak H, Ioannidis JP, Argiles A, Attwood TK, Bongcam-Rudloff E, et al. (2012) Implementation of proteomic biomarkers: making it work. Eur J Clin Invest 42: 1027–1036.
    1. Mischak H, Vlahou A, Righetti PG, Calvete JJ (2014) Putting value in biomarker research and reporting. J Proteomics 96: A1–A3.
    1. Unterwurzacher I, Koal T, Bonn GK, Weinberger KM, Ramsay SL (2008) Rapid sample preparation and simultaneous quantitation of prostaglandins and lipoxygenase derived fatty acid metabolites by liquid chromatography-mass spectrometry from small sample volumes. Clin Chem Lab Med 46: 1589–1597.
    1. Eibl G, Bernardo K, Koal T, Ramsay SL, Weinberger KM, et al. (2008) Isotope correction of mass spectrometry profiles. Rapid Commun Mass Spectrom 22: 2248–2252.
    1. Haubitz M, Good DM, Woywodt A, Haller H, Rupprecht H, et al. (2009) Identification and validation of urinary biomarkers for differential diagnosis and dvaluation of therapeutic intervention in ANCA associated vasculitis. Mol Cell Proteomics 8: 2296–2307.
    1. Mischak H, Vlahou A, Ioannidis JP (2013) Technical aspects and inter-laboratory variability in native peptide profiling: The CE-MS experience. Clin Biochem 46: 432–443.
    1. Neuhoff N, Kaiser T, Wittke S, Krebs R, Pitt A, et al. (2004) Mass spectrometry for the detection of differentially expressed proteins: a comparison of surface-enhanced laser desorption/ionization and capillary electrophoresis/mass spectrometry. Rapid Communications in Mass Spectrometry 18: 149–156.
    1. Jantos-Siwy J, Schiffer E, Brand K, Schumann G, Rossing K, et al. (2009) Quantitative Urinary Proteome Analysis for Biomarker Evaluation in Chronic Kidney Disease. J Proteome Res 8: 268–281.
    1. Dakna M, He Z, Yu WC, Mischak H, Kolch W (2009) Technical, bioinformatical and statistical aspects of liquid chromatography-mass spectrometry (LC-MS) and capillary electrophoresis-mass spectrometry (CE-MS) based clinical proteomics: a critical assessment. J Chromatogr B Analyt Technol Biomed Life Sci 877: 1250–1258.
    1. Benjamini Y, Hochberg Y (1995) Controlling the false discovery rate: a practical and powerful approach to multiple testing. J Royal Stat Soc B (Methodological) 57: 125–133.
    1. Girolami M, Mischak H, Krebs R (2006) Analysis of complex, multidimensional datasets. Drug Discov Today: Technologies 3: 13–19.
    1. Yang ZR (2004) Biological applications of support vector machines. Brief Bioinform 5: 328–338.
    1. R Development Core Team (2008) R: A language and environment for statistical computing. Vienna, Austria: R Foundation for Statistical Computing.
    1. Duranton F, Lundin U, Gayrard N, Mischak H, Aparicio M, et al... (2013) Plasma and Urinary Amino Acid Metabolomic Profiling in Patients with Different Levels of Kidney Function. Clin J Am Soc Nephrol.
    1. Prajczer S, Heidenreich U, Pfaller W, Kotanko P, Lhotta K, et al. (2010) Evidence for a role of uromodulin in chronic kidney disease progression. Nephrol Dial Transplant 25: 1896–1903.
    1. Zhou J, Chen Y, Liu Y, Shi S, Wang S, et al. (2013) Urinary uromodulin excretion predicts progression of chronic kidney disease resulting from IgA nephropathy. PLoS ONE 8: e71023.
    1. Neilson EG (2006) Mechanisms of disease: Fibroblasts—a new look at an old problem. Nat Clin Pract Nephrol 2: 101–108.
    1. Boor P, Ostendorf T, Floege J (2010) Renal fibrosis: novel insights into mechanisms and therapeutic targets. Nat Rev Nephrol 6: 643–656.
    1. Cheng S, Pollock AS, Mahimkar R, Olson JL, Lovett DH (2006) Matrix metalloproteinase 2 and basement membrane integrity: a unifying mechanism for progressive renal injury. FASEB J 20: 1898–1900.
    1. Rodriguez-Iturbe B, Ferrebuz A, Vanegas V, Quiroz Y, Espinoza F, et al. (2005) Early treatment with cGMP phosphodiesterase inhibitor ameliorates progression of renal damage. Kidney Int 68: 2131–2142.
    1. Ruster C, Wolf G (2006) Renin-angiotensin-aldosterone system and progression of renal disease. J Am Soc Nephrol 17: 2985–2991.
    1. Vogl T, Ludwig S, Goebeler M, Strey A, Thorey IS, et al. (2004) MRP8 and MRP14 control microtubule reorganization during transendothelial migration of phagocytes. Blood 104: 4260–4268.
    1. Boger RH, Zoccali C (2003) ADMA: a novel risk factor that explains excess cardiovascular event rate in patients with end-stage renal disease. Atheroscler Suppl 4: 23–28.
    1. Duranton F, Lundin U, Gayrard N, Mischak H, Aparicio M, et al. (2014) Plasma and urinary amino Acid metabolomic profiling in patients with different levels of kidney function. Clin J Am Soc Nephrol 9: 37–45.
    1. Goek ON, Prehn C, Sekula P, Romisch-Margl W, Doring A, et al. (2013) Metabolites associate with kidney function decline and incident chronic kidney disease in the general population. Nephrol Dial Transplant 28: 2131–2138.
    1. Shah VO, Townsend RR, Feldman HI, Pappan KL, Kensicki E, et al. (2013) Plasma metabolomic profiles in different stages of CKD. Clin J Am Soc Nephrol 8: 363–370.
    1. Duranton F, Lundin U, Gayrard N, Mischak H, Aparicio M, et al... (2013) Plasma and Urinary Amino Acid Metabolomic Profiling in Patients with Different Levels of Kidney Function. Clin J Am Soc Nephrol.
    1. Tsikas D, Boger RH, Sandmann J, Bode-Boger SM, Frolich JC (2000) Endogenous nitric oxide synthase inhibitors are responsible for the L-arginine paradox. FEBS Lett 478: 1–3.
    1. Kielstein JT, Boger RH, Bode-Boger SM, Schaffer J, Barbey M, et al. (1999) Asymmetric dimethylarginine plasma concentrations differ in patients with end-stage renal disease: relationship to treatment method and atherosclerotic disease. J Am Soc Nephrol 10: 594–600.
    1. Kielstein JT, Donnerstag F, Gasper S, Menne J, Kielstein A, et al. (2006) ADMA increases arterial stiffness and decreases cerebral blood flow in humans. Stroke 37: 2024–2029.
    1. Fouque D, Holt S, Guebre-Egziabher F, Nakamura K, Vianey-Saban C, et al. (2006) Relationship between serum carnitine, acylcarnitines, and renal function in patients with chronic renal disease. J Ren Nutr 16: 125–131.
    1. Jin K, Norris K, Vaziri ND (2013) Dysregulation of hepatic fatty acid metabolism in chronic kidney disease. Nephrol Dial Transplant 28: 313–320.
    1. Vaziri ND (2009) Causes of dysregulation of lipid metabolism in chronic renal failure. Semin Dial 22: 644–651.
    1. Drosatos K, Schulze PC (2013) Cardiac lipotoxicity: molecular pathways and therapeutic implications. Curr Heart Fail Rep 10: 109–121.
    1. Guebre-Egziabher F, Alix PM, Koppe L, Pelletier CC, Kalbacher E, et al. (2013) Ectopic lipid accumulation: A potential cause for metabolic disturbances and a contributor to the alteration of kidney function. Biochimie 95: 1971–1979.
    1. Li LO, Klett EL, Coleman RA (2010) Acyl-CoA synthesis, lipid metabolism and lipotoxicity. Biochim Biophys Acta 1801: 246–251.
    1. Rebouche CJ (2004) Kinetics, pharmacokinetics, and regulation of L-carnitine and acetyl-L-carnitine metabolism. Ann N Y Acad Sci 1033: 30–41.
    1. Schmidt-Sommerfeld E, Penn D, Bieber LL, Kerner J, Rossi TM, et al. (1990) Carnitine ester excretion in pediatric patients receiving parenteral nutrition. Pediatr Res 28: 158–165.
    1. Reuter SE, Evans AM (2012) Carnitine and acylcarnitines: pharmacokinetic, pharmacological and clinical aspects. Clin Pharmacokinet 51: 553–572.
    1. Wanner C, Forstner-Wanner S, Rossle C, Furst P, Schollmeyer P, et al... (1987) Carnitine metabolism in patients with chronic renal failure: effect of L-carnitine supplementation. Kidney Int Suppl 22: S132–S135.
    1. Tankiewicz A, Pawlak D, Topczewska-Bruns J, Buczko W (2003) Kidney and liver kynurenine pathway enzymes in chronic renal failure. Adv Exp Med Biol 527: 409–414.
    1. Duranton F, Lundin U, Gayrard N, Mischak H, Aparicio M, et al... (2013) Plasma and Urinary Amino Acid Metabolomic Profiling in Patients with Different Levels of Kidney Function. Clin J Am Soc Nephrol.
    1. Pawlak K, Tankiewicz J, Mysliwiec M, Pawlak D (2009) Tissue factor/its pathway inhibitor system and kynurenines in chronic kidney disease patients on conservative treatment. Blood Coagul Fibrinolysis 20: 590–594.
    1. Pawlak K, Domaniewski T, Mysliwiec M, Pawlak D (2009) The kynurenines are associated with oxidative stress, inflammation and the prevalence of cardiovascular disease in patients with end-stage renal disease. Atherosclerosis 204: 309–314.

Source: PubMed

3
Sottoscrivi