Strategy and rationale for urine collection protocols employed in the NEPTUNE study

Marie C Hogan, John C Lieske, Chrysta C Lienczewski, Lisa L Nesbitt, Larysa T Wickman, Christina M Heyer, Peter C Harris, Christopher J Ward, Jamie L Sundsbak, Luca Manganelli, Wenjun Ju, Jeffrey B Kopp, Peter J Nelson, Sharon G Adler, Heather N Reich, Lawrence B Holzmann, Matthias Kretzler, Markus Bitzer, Marie C Hogan, John C Lieske, Chrysta C Lienczewski, Lisa L Nesbitt, Larysa T Wickman, Christina M Heyer, Peter C Harris, Christopher J Ward, Jamie L Sundsbak, Luca Manganelli, Wenjun Ju, Jeffrey B Kopp, Peter J Nelson, Sharon G Adler, Heather N Reich, Lawrence B Holzmann, Matthias Kretzler, Markus Bitzer

Abstract

Background: Glomerular diseases are potentially fatal, requiring aggressive interventions and close monitoring. Urine is a readily-accessible body fluid enriched in molecular signatures from the kidney and therefore particularly suited for routine clinical analysis as well as development of non-invasive biomarkers for glomerular diseases.

Methods: The Nephrotic Syndrome Study Network (NEPTUNE; ClinicalTrials.gov Identifier NCT01209000) is a North American multicenter collaborative consortium established to develop a translational research infrastructure for nephrotic syndrome. This includes standardized urine collections across all participating centers for the purpose of discovering non-invasive biomarkers for patients with nephrotic syndrome due to minimal change disease, focal segmental glomerulosclerosis, and membranous nephropathy. Here we describe the organization and methods of urine procurement and banking procedures in NEPTUNE.

Results: We discuss the rationale for urine collection and storage conditions, and demonstrate the performance of three experimental analytes (neutrophil gelatinase-associated lipocalin [NGAL], retinol binding globulin, and alpha-1 microglobulin) under these conditions with and without urine preservatives (thymol, toluene, and boric acid). We also demonstrate the quality of RNA and protein collected from the urine cellular pellet and exosomes.

Conclusions: The urine collection protocol in NEPTUNE allows robust detection of a wide range of proteins and RNAs from urine supernatant and pellets collected longitudinally from each patient over 5 years. Combined with the detailed clinical and histopathologic data, this provides a unique resource for exploration and validation of new or accepted markers of glomerular diseases.

Trial registration: ClinicalTrials.gov Identifier NCT01209000.

Figures

Fig. 1
Fig. 1
a NEPTUNE spot urine sample processing: Sample 1: 4 × 2 mL cryovials whole unprocessed “raw” urine (SU). Sample 2: 4 × 2 mL samples with sodium azide additive (AS). Supernatant after low speed centrifugation is saved in cryovials with 20 μL 100 mM sodium azide (12 mL × 4). Sodium azide has anti-bacterial properties. Sample 3: 4 cryovials with proteinase inhibitor (PI). Sample 4: 2 sets of pellet from this are stored in RNAlater obtained by centrifugation at 1000 g × 12 min in a tabletop centrifuge (AP-E). Sample 5: 4 × 2 mL from the PI sample. 2 sets of pellet from this are stored in in RNAlater obtained by centrifugation at 1000 g x 12 min in a tabletop centrifuge (AQ-Q). All samples are stored on ice in transport and during collection. All samples are frozen as soon possible at −80 °C. Cryovial final volume stored is 1.6 mL urine. b NEPTUNE 24-h urine sample processing. One subaliquot 40 mL whole urine (inverted x 3) frozen at −80 °C immediately (U1). 1 subaliquot (40 mL) of whole urine with protease inhibitor (6 μL; Sigma P1860) (UQ1); 5 × 5 mL cryovials whole urine (U-24)
Fig. 2
Fig. 2
Immunoassay of soluble proteins. a Neutrophil gelatinase-associated lipocalin (NGAL) (b). Alpha-1 microglobulin (AMBP). c Retinol-binding globulin. All 3 were stable at room temperature, 4 °C, and −20 °C in urine (shown in this figure) without preservatives and also with toluene, thymol, and boric acid. None were stable with acetic acid, 6 N HCl, or 6 N HNO3 (Additional file 2: Table S3). Median and interquartile ranges are shown
Fig. 3
Fig. 3
Western blot of exosome pellets. Exosomal proteins of interest including podocalyxin (PODXL), fibrocystin (FIBRO) and polycystin 1 (PC1) were examined by Western blot after storage and compared with freshly-isolated exosomes. These proteins were best preserved by the Sigma protease cocktail. a Polycystin 1 detection by Western blot was stable for 1 week at room temperature after exosomes were isolated and stored in 0.01 % sodium azide (n = 7). However, polycystin 1 was not detected well from exosomes isolated after urine was frozen.at -80C probably because of precipitation issues and unlikely due to loss of protease activity. Effects of preservatives on Western blot of exosome pellet proteins PODXL, scramblase (PLSCR1), FIBRO, and smoothened (SMO). b Lane 1, Fresh, no preservatives; Lane 2, Fresh, no preservatives; Lane 3, −80 °C Frozen/Thawed; Lane 4, −80 °C Frozen/Thawed. c Comparison of varied amounts of protease inhibitors on exosome protein detection by Western blot. Initial assessment and following 6 months storage at −80 °C. Lane 1, 4.8 μL. Sigma protease inhibitor; Lane 2, 4.0 μL Sigma protease inhibitor; Lane 3, 1:100 Roche Complete tablet; Lane 4, −80 °C Frozen/Thawed; Lane 5, Fresh, no preservatives. d Exosomal Podocalyxin fared well at RT and at 4 °C. Sodium azide did not affect their survival. e Analysis of exosomes extracted from frozen (−80 °C) raw urine stored for 12 months

References

    1. Meyers CM, Geanacopoulos M, Holzman LB, Salant DJ. Glomerular disease workshop. J Am Soc Nephrol. 2005;16:3472–6. doi: 10.1681/ASN.2005090899.
    1. Standard Protocol for Urine Collection and Storage []. Access date 11/15/2015.
    1. Elliott P, Peakman TC. The UK Biobank sample handling and storage protocol for the collection, processing and archiving of human blood and urine. Int J Epidemiol. 2008;37:234–44. doi: 10.1093/ije/dym276.
    1. Thongboonkerd V. Practical points in urinary proteomics. J Proteome Res. 2007;6:3881–90. doi: 10.1021/pr070328s.
    1. Thongboonkerd V, Chutipongtanate S, Kanlaya R. Systematic evaluation of sample preparation methods for gel-based human urinary proteomics: quantity, quality, and variability. J Proteome Res. 2006;5:183–91. doi: 10.1021/pr0502525.
    1. Adachi J, Kumar C, Zhang Y, Olsen JV, Mann M. The human urinary proteome contains more than 1500 proteins, including a large proportion of membrane proteins. Genome Biol. 2006;7:R80. doi: 10.1186/gb-2006-7-9-r80.
    1. Nephrotic Syndrome Study Network (“NEPTUNE”) []. Access date 11/15/2015.
    1. Devarajan P. Neutrophil gelatinase-associated lipocalin (NGAL): a new marker of kidney disease. Scand J Clin Lab Invest Suppl. 2008;68:89–94. doi: 10.1080/00365510802150158.
    1. Youssef DM, El-Shal AA. Urine neutrophil gelatinase-associated lipocalin and kidney injury in children with focal segmental glomerulosclerosis. Iran J Kidney Dis. 2012;6:355–60.
    1. Bennett MR, Piyaphanee N, Czech K, Mitsnefes M, Devarajan P. NGAL distinguishes steroid sensitivity in idiopathic nephrotic syndrome. Pediatr Nephrol. 2012;27:807–12. doi: 10.1007/s00467-011-2075-7.
    1. Weber MH, Verwiebe R. Alpha 1-microglobulin (protein HC): features of a promising indicator of proximal tubular dysfunction. Eur J Clin Chem Clin Biochem. 1992;30:683–91.
    1. Bernard AM, Vyskocil AA, Mahieu P, Lauwerys RR. Assessment of urinary retinol-binding protein as an index of proximal tubular injury. Clin Chem. 1987;33:775–9.
    1. Zhang Q, Zeng C, Cheng Z, Xie K, Zhang J, Liu Z. Primary focal segmental glomerulosclerosis in nephrotic patients: common complications and risk factors. J Nephrol. 2012;25:679–88. doi: 10.5301/jn.5000040.
    1. Bazzi C, Petrini C, Rizza V, Arrigo G, Beltrame A, Pisano L, et al. Urinary excretion of IgG and [alpha] 1-microglobulin predicts clinical course better than extent of proteinuria in membranous nephropathy. Am J Kidney Dis. 2001;38:240–8. doi: 10.1053/ajkd.2001.26080.
    1. Varghese SA, Powell TB, Budisavljevic MN, Oates JC, Raymond JR, Almeida JS, et al. Urine biomarkers predict the cause of glomerular disease. J Am Soc Nephrol. 2007;18:913–22. doi: 10.1681/ASN.2006070767.
    1. Kershaw DB, Beck SG, Wharram BL, Wiggins JE, Goyal M, Thomas PE, et al. Molecular cloning and characterization of human podocalyxin-like protein. Orthologous relationship to rabbit PCLP1 and rat podocalyxin. J Biol Chem. 1997;272:15708–14. doi: 10.1074/jbc.272.25.15708.
    1. Habara P, Mareckova H, Sopkova Z, Malickova K, Zivorova D, Zima T, et al. A novel method for the estimation of podocyte injury: podocalyxin-positive elements in urine. Folia Biol (Praha) 2008;54:162–7.
    1. Kanno K, Kawachi H, Uchida Y, Hara M, Shimizu F, Uchiyama M. Urinary sediment podocalyxin in children with glomerular diseases. Nephron Clin Pract. 2003;95:c91–9. doi: 10.1159/000074322.
    1. Ward CJ, Hogan MC, Rossetti S, Walker D, Sneddon T, Wang X, et al. The gene mutated in autosomal recessive polycystic kidney disease encodes a large, receptor-like protein. Nat Genet. 2002;30:259–69. doi: 10.1038/ng833.
    1. Hughes J, Ward CJ, Peral B, Aspinwall R, Clark K, San Millan JL, et al. The polycystic kidney disease 1 (PKD1) gene encodes a novel protein with multiple cell recognition domains. Nat Genet. 1995;10:151–60. doi: 10.1038/ng0695-151.
    1. Corbit KC, Aanstad P, Singla V, Norman AR, Stainier DY, Reiter JF. Vertebrate Smoothened functions at the primary cilium. Nature. 2005;437:1018–21. doi: 10.1038/nature04117.
    1. Hogan MC, Manganelli L, Woollard JR, Masyuk AI, Masyuk TV, Tammachote R, et al. Characterization of PKD protein-positive exosome-like vesicles. J Am Soc Nephrol. 2009;20:278–88. doi: 10.1681/ASN.2008060564.
    1. Sato Y, Wharram BL, Lee SK, Wickman L, Goyal M, Venkatareddy M, et al. Urine podocyte mRNAs mark progression of renal disease. J Am Soc Nephrol. 2009;20:1041–52. doi: 10.1681/ASN.2007121328.
    1. Wishart DS, Tzur D, Knox C, Eisner R, Guo AC, Young N, et al. HMDB: the human metabolome database. Nucleic Acids Res. 2007;35:D521–6. doi: 10.1093/nar/gkl923.
    1. Kuhara T. Gas chromatographic-mass spectrometric urinary metabolome analysis to study mutations of inborn errors of metabolism. Mass Spectrom Rev. 2005;24:814–27. doi: 10.1002/mas.20038.
    1. Dear JW, Street JM, Bailey MA. Urinary exosomes: a reservoir for biomarker discovery and potential mediators of intra-renal signaling. Proteomics. 2012;13:1572–80. doi: 10.1002/pmic.201200285.
    1. Nagaraj N, Mann M. Quantitative analysis of the intra- and inter-individual variability of the normal urinary proteome. J Proteome Res. 2011;10:637–45. doi: 10.1021/pr100835s.
    1. Schinstock CA, Semret MH, Wagner SJ, Borland TM, Bryant SC, Kashani KB, et al. Urinalysis is more specific and urinary neutrophil gelatinase-associated lipocalin is more sensitive for early detection of acute kidney injury. Nephrol Dial Transplant. 2013;28:1175–85. doi: 10.1093/ndt/gfs127.
    1. Bitzer M, Ju W, Jing X, Zavadil J. Quantitative analysis of miRNA expression in epithelial cells and tissues. Methods Mol Biol. 2012;820:55–70. doi: 10.1007/978-1-61779-439-1_4.
    1. Best Practices for Sample Storage: Urine as a Paradigm Workshop on Urine Biospecimen Handling Meeting. In: NIH Workshop: February 22–23, 2010 2010; Bethesda, MD; 2010.
    1. Human Kidney and Urine Proteome Project []. Access date 11/15/2015.
    1. Smets PMY, Meyer E, Maddens B, Duchateau L, Daminet S. Effect of sampling method and storage conditions on albumin, retinol-binding protein, and N-acetyl-β-D-glucosaminidase concentrations in canine urine samples. J Vet Diagn Invest. 2010;22:896–902. doi: 10.1177/104063871002200607.
    1. Chen C, Ward CJ, Hopp K, Harris PC, Hogan MC. Systematic analysis of microRNAs in urine exosome-like vesicles and correlation with Autosomal Dominant Polycystic Kidney Disease (ADPKD). J Am Soc Nephrol. 2012;246A.
    1. Munch EM, Harris RA, Mohammad M, Benham AL, Pejerrey SM, Showalter L, et al. Transcriptome profiling of microRNA by next-Gen deep sequencing reveals known and novel miRNA species in the lipid fraction of human breast milk. PLoS One. 2013;8:e50564. doi: 10.1371/journal.pone.0050564.
    1. Alvarez ML, Khosroheidari M, Kanchi Ravi R, Distefano JK. Comparison of protein, microRNA, and mRNA yields using different methods of urinary exosome isolation for the discovery of kidney disease biomarkers. Kidney Int. 2012.
    1. Keller S, Ridinger J, Rupp AK, Janssen JW, Altevogt P. Body fluid derived exosomes as a novel template for clinical diagnostics. J Transl Med. 2011;9:86. doi: 10.1186/1479-5876-9-86.
    1. Koh W, Sheng CT, Tan B, Lee QY, Kuznetsov V, Kiang LS et al. Analysis of deep sequencing microRNA expression profile from human embryonic stem cells derived mesenchymal stem cells reveals possible role of let-7 microRNA family in downstream targeting of hepatic nuclear factor 4 alpha. BMC Genomics. 2010, 11 Suppl 1:S6.
    1. Rabinowits G, Gercel-Taylor C, Day JM, Taylor DD, Kloecker GH. Exosomal microRNA: a diagnostic marker for lung cancer. Clin Lung Cancer. 2009;10:42–6. doi: 10.3816/CLC.2009.n.006.
    1. Eirin A, Irazabal MV, Gertz MA, Dispenzieri A, Lacy MQ, Kumar S, et al. Clinical features of patients with immunoglobulin light chain amyloidosis (AL) with vascular-limited deposition in the kidney. Nephrol Dial Transplant. 2012;27:1097–101. doi: 10.1093/ndt/gfr381.
    1. Gudehithlu KP, Pegoraro AA, Dunea G, Arruda JAL, Singh AK. Degradation of albumin by the renal proximal tubule cells and the subsequent fate of its fragments. Kidney Int. 2004;65:2113–22. doi: 10.1111/j.1523-1755.2004.00633.x.
    1. Hogan MC, Johnson KL, Zenka RM, Charlesworth MC, Madden BJ, Mahoney DW, et al. Subfractionation, characterization, and in-depth proteomic analysis of glomerular membrane vesicles in human urine. Kidney Int. 2014;85:1225–37. doi: 10.1038/ki.2013.422.
    1. Gonzales PA, Pisitkun T, Hoffert JD, Tchapyjnikov D, Star RA, Kleta R, et al. Large-scale proteomics and phosphoproteomics of urinary exosomes. J Am Soc Nephrol. 2009;20:363–79. doi: 10.1681/ASN.2008040406.
    1. Gonzales P, Pisitkun T, Knepper MA. Urinary exosomes: is there a future? Nephrol Dial Transplant. 2008;23:1799–801. doi: 10.1093/ndt/gfn058.
    1. Wang Z, Hill S, Luther JM, Hachey DL, Schey KL. Proteomic analysis of urine exosomes by multidimensional protein identification technology (MudPIT) Proteomics. 2012;12:329–38. doi: 10.1002/pmic.201100477.
    1. Cheruvanky A, Zhou H, Pisitkun T, Kopp JB, Knepper MA, Yuen PS, et al. Rapid isolation of urinary exosomal biomarkers using a nanomembrane ultrafiltration concentrator. Am J Physiol Renal Physiol. 2007;292:F1657–61. doi: 10.1152/ajprenal.00434.2006.
    1. Gonzales PA, Zhou H, Pisitkun T, Wang NS, Star RA, Knepper MA, et al. Isolation and purification of exosomes in urine. Methods Mol Biol. 2010;641:89–99. doi: 10.1007/978-1-60761-711-2_6.
    1. Merchant ML, Powell DW, Wilkey DW, Cummins TD, Deegens JK, Rood IM, et al. Microfiltration isolation of human urinary exosomes for characterization by MS. Proteom Clin Appl. 2010;4:84–96. doi: 10.1002/prca.200800093.
    1. Raj DAA, Fiume I, Capasso G, Pocsfalvi G. A multiplex quantitative proteomics strategy for protein biomarker studies in urinary exosomes. Kidney Int. 2012;81:1263–72. doi: 10.1038/ki.2012.25.
    1. Pisitkun T, Shen RF, Knepper MA. Identification and proteomic profiling of exosomes in human urine. Proc Natl Acad Sci U S A. 2004;101:13368–73. doi: 10.1073/pnas.0403453101.
    1. Rood IM, Deegens JKJ, Merchant ML, Tamboer WPM, Wilkey DW, Wetzels JFM, et al. Comparison of three methods for isolation of urinary microvesicles to identify biomarkers of nephrotic syndrome. Kidney Int. 2010;78:810–6. doi: 10.1038/ki.2010.262.

Source: PubMed

3
Se inscrever