A comprehensive analysis and annotation of human normal urinary proteome

Mindi Zhao, Menglin Li, Yehong Yang, Zhengguang Guo, Ying Sun, Chen Shao, Mingxi Li, Wei Sun, Youhe Gao, Mindi Zhao, Menglin Li, Yehong Yang, Zhengguang Guo, Ying Sun, Chen Shao, Mingxi Li, Wei Sun, Youhe Gao

Abstract

Biomarkers are measurable changes associated with the disease. Urine can reflect the changes of the body while blood is under control of the homeostatic mechanisms; thus, urine is considered an important source for early and sensitive disease biomarker discovery. A comprehensive profile of the urinary proteome will provide a basic understanding of urinary proteins. In this paper, we present an in-depth analysis of the urinary proteome based on different separation strategies, including direct one dimensional liquid chromatography-tandem mass spectrometry (LC/MS/MS), two dimensional LC/MS/MS, and gel-eluted liquid fraction entrapment electrophoresis/liquid-phase isoelectric focusing followed by two dimensional LC/MS/MS. A total of 6085 proteins were identified in healthy urine, of which 2001 were not reported in previous studies and the concentrations of 2571 proteins were estimated (spanning a magnitude of 106) with an intensity-based absolute quantification algorithm. The urinary proteins were annotated by their tissue distribution. Detailed information can be accessed at the "Human Urine Proteome Database" (www.urimarker.com/urine).

Conflict of interest statement

The authors declare that they have no competing interests.

Figures

Figure 1
Figure 1
The workflow of urinary proteome analysis. Pooled urine from 24 humans was analyzed using three separation strategies. 1D: Urinary peptides were directly analyzed via 1DLC/MS/MS without fractionation. 2D: Urinary peptides were analyzed via offline RPLC and 1DLC/MS/MS. 3D: Urinary proteins were first fractionated by GELFrEE/LP-IEF prior to offline RPLC. A total of 383 fractions were analyzed by LC/MS/MS using high-resolution TripleTOF 5600 MS. A urine proteome database was then constructed based on bioinformatics analyses.
Figure 2
Figure 2
The results from three separation strategies. (A) A Coomassie-stained Bis-Tris gel image of 12 GELFrEE fractions over a broad mass range. (B) Coomassie-stained Bis-Tris gel image of 10 LP-IEF fractions over a pI range from 3 to 10. (C) Venn diagram of proteins identified by three separation strategies. (D) Venn diagram of proteins identified from this study as well as previous urine and exosome proteome studies. (E) Comparative analysis of the urine, kidney and plasma proteome.
Figure 3
Figure 3
Quantitative analysis of urinary proteins by the iBAQ method. (A) The relative expression and concentrations of 2,571 proteins in the 2D analysis were estimated by iBAQ. The left y-axis represents relative abundance, and the right y axis represents estimated concentration (pg/mL). (1) ALB: albumin; UROM: uromodulin, the two most abundant proteins. (2) RARS: arginine-tRNA ligase, the least abundant protein in 2D analysis; (3) RNASE 6: ribonuclease K6, the least abundant protein in 1D analysis. (B) Correlation plot between estimated concentrations and immunoassays results.
Figure 4
Figure 4
Cellular component and canonical pathway analyses of three separation groups. (A) Cellular component analysis of the three groups. (B) The top 10 canonical pathways from the three groups. The y-axis denotes the negative log of the p value.
Figure 5
Figure 5
Tissue distribution of urinary proteins at protein level. (A) Urinary proteome distributions across 44 tissues. The numbers in the bracket denote the number of highly expressed proteins of the tissue detected in urine. (B) The distribution of tissue-related proteins and the corresponding separation strategy for top ten tissues.
Figure 6
Figure 6
An overview of the human urinary proteome database. (A) The protein level results include the unique peptide count, total peptide count and relative quantitation and estimated concentration. Proteins are linked to the UniProt website by clicking the accessions. (B) The peptide level results include peptide sequences and observed m/z values. (C) The database provides the experimental pI and MW distribution of all identified proteins. (D) The “MW-PI” section provides a succinct figure summarizing the theoretical MW and pI information for each protein. (E) Biomarker application of all identified proteins.

References

    1. Casey, H. W., Ayers, K. N. & Robinson, F. In Pathology of laboratory animals 115–173 (Springer, 1978).
    1. Gao Y. Urine-an untapped goldmine for biomarker discovery? Science China. Life sciences. 2013;56:1145–1146. doi: 10.1007/s11427-013-4574-1.
    1. Sun W, et al. Human urine proteome analysis by three separation approaches. Proteomics. 2005;5:4994–5001. doi: 10.1002/pmic.200401334.
    1. Ho J, et al. Mass spectrometry-based proteomic analysis of urine in acute kidney injury following cardiopulmonary bypass: a nested case-control study. American journal of kidney diseases: the official journal of the National Kidney Foundation. 2009;53:584–595. doi: 10.1053/j.ajkd.2008.10.037.
    1. Orenes-Pinero E, et al. Searching urinary tumor markers for bladder cancer using a two-dimensional differential gel electrophoresis (2D-DIGE) approach. Journal of proteome research. 2007;6:4440–4448. doi: 10.1021/pr070368w.
    1. Zubiri I, et al. Diabetic nephropathy induces changes in the proteome of human urinary exosomes as revealed by label-free comparative analysis. Journal of proteomics. 2014;96:92–102. doi: 10.1016/j.jprot.2013.10.037.
    1. Thuijls G, et al. Early diagnosis of intestinal ischemia using urinary and plasma fatty acid binding proteins. Annals of surgery. 2011;253:303–308. doi: 10.1097/SLA.0b013e318207a767.
    1. Liu L, et al. Evaluation of urinary S100B protein level and lactate/creatinine ratio for early diagnosis and prognostic prediction of neonatal hypoxic-ischemic encephalopathy. Neonatology. 2010;97:41–44. doi: 10.1159/000227292.
    1. Chugh S, et al. Pilot study identifying myosin heavy chain 7, desmin, insulin-like growth factor 7, and annexin A2 as circulating biomarkers of human heart failure. Proteomics. 2013;13:2324–2334. doi: 10.1002/pmic.201200455.
    1. Davis, M. T. et al. Towards defining the urinary proteome using liquid chromatography-tandem mass spectrometry. II. Limitations of complex mixture analyses. Proteomics1, 108–117, doi:10.1002/1615-9861(200101)1:1<108::AID-PROT108>;2-5 (2001).
    1. Spahr, C. S. et al. Towards defining the urinary proteome using liquid chromatography-tandem mass spectrometry. I. Profiling an unfractionated tryptic digest. Proteomics1, 93–107, doi:10.1002/1615-9861(200101)1:1<93::AID-PROT93>;2-3 (2001).
    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 biology. 2006;7:R80. doi: 10.1186/gb-2006-7-9-r80.
    1. Marimuthu A, et al. A comprehensive map of the human urinary proteome. Journal of proteome research. 2011;10:2734–2743. doi: 10.1021/pr2003038.
    1. Li QR, et al. A comprehensive and non-prefractionation on the protein level approach for the human urinary proteome: touching phosphorylation in urine. Rapid communications in mass spectrometry: RCM. 2010;24:823–832. doi: 10.1002/rcm.4441.
    1. Zheng J, Liu L, Wang J, Jin Q. Urinary proteomic and non-prefractionation quantitative phosphoproteomic analysis during pregnancy and non-pregnancy. BMC genomics. 2013;14:777. doi: 10.1186/1471-2164-14-777.
    1. Santucci L, et al. From hundreds to thousands: Widening the normal human Urinome (1) Journal of proteomics. 2014;112C:53–62.
    1. Desiere F, et al. The PeptideAtlas project. Nucleic acids research. 2006;34:D655–658. doi: 10.1093/nar/gkj040.
    1. Kim MS, et al. A draft map of the human proteome. Nature. 2014;509:575–581. doi: 10.1038/nature13302.
    1. Wilhelm M, et al. Mass-spectrometry-based draft of the human proteome. Nature. 2014;509:582–587. doi: 10.1038/nature13319.
    1. Consortium EP. An integrated encyclopedia of DNA elements in the human genome. Nature. 2012;489:57–74. doi: 10.1038/nature11247.
    1. Tran JC, Doucette AA. Gel-eluted liquid fraction entrapment electrophoresis: an electrophoretic method for broad molecular weight range proteome separation. Analytical chemistry. 2008;80:1568–1573. doi: 10.1021/ac702197w.
    1. Ahmed FE. Sample preparation and fractionation for proteome analysis and cancer biomarker discovery by mass spectrometry. Journal of separation science. 2009;32:771–798. doi: 10.1002/jssc.200900014.
    1. Gonzales PA, et al. Large-scale proteomics and phosphoproteomics of urinary exosomes. Journal of the American Society of Nephrology: JASN. 2009;20:363–379. doi: 10.1681/ASN.2008040406.
    1. Wang Z, Hill S, Luther JM, Hachey DL, Schey KL. Proteomic analysis of urine exosomes by multidimensional protein identification technology (MudPIT) Proteomics. 2012;12:329–338. doi: 10.1002/pmic.201100477.
    1. Hogan MC, et al. Subfractionation, characterization, and in-depth proteomic analysis of glomerular membrane vesicles in human urine. Kidney international. 2014;85:1225–1237. doi: 10.1038/ki.2013.422.
    1. Huang da W, Sherman BT, Lempicki RA. Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources. Nature protocols. 2009;4:44–57. doi: 10.1038/nprot.2008.211.
    1. Huang da W, Sherman BT, Lempicki RA. Bioinformatics enrichment tools: paths toward the comprehensive functional analysis of large gene lists. Nucleic acids research. 2009;37:1–13. doi: 10.1093/nar/gkn923.
    1. Jia L, et al. An attempt to understand kidney’s protein handling function by comparing plasma and urine proteomes. PloS one. 2009;4:e5146. doi: 10.1371/journal.pone.0005146.
    1. Farrah T, et al. State of the human proteome in 2013 as viewed through PeptideAtlas: comparing the kidney, urine, and plasma proteomes for the biology- and disease-driven Human Proteome Project. Journal of proteome research. 2014;13:60–75. doi: 10.1021/pr4010037.
    1. Pieper R, et al. Characterization of the human urinary proteome: a method for high-resolution display of urinary proteins on two-dimensional electrophoresis gels with a yield of nearly 1400 distinct protein spots. Proteomics. 2004;4:1159–1174. doi: 10.1002/pmic.200300661.
    1. Geiger T, Wehner A, Schaab C, Cox J, Mann M. Comparative proteomic analysis of eleven common cell lines reveals ubiquitous but varying expression of most proteins. Molecular & cellular proteomics: MCP. 2012;11:M111 014050. doi: 10.1074/mcp.M111.014050.
    1. Nagaraj N, Mann M. Quantitative analysis of the intra- and inter-individual variability of the normal urinary proteome. Journal of proteome research. 2011;10:637–645. doi: 10.1021/pr100835s.
    1. Dyer AR, et al. Evaluation of measures of urinary albumin excretion in epidemiologic studies. American journal of epidemiology. 2004;160:1122–1131. doi: 10.1093/aje/kwh326.
    1. Nolen BM, et al. An extensive targeted proteomic analysis of disease-related protein biomarkers in urine from healthy donors. PloS one. 2013;8:e63368. doi: 10.1371/journal.pone.0063368.
    1. Kentsis A, et al. Discovery and validation of urine markers of acute pediatric appendicitis using high-accuracy mass spectrometry. Annals of emergency medicine. 2010;55:62–70 e64. doi: 10.1016/j.annemergmed.2009.04.020.
    1. Shao C, Wang Y, Gao Y. Applications of urinary proteomics in biomarker discovery. Science China. Life sciences. 2011;54:409–417. doi: 10.1007/s11427-011-4162-1.
    1. Suh JH, Miner JH. The glomerular basement membrane as a barrier to albumin. Nature reviews. Nephrology. 2013;9:470–477. doi: 10.1038/nrneph.2013.109.
    1. Welsh GI, Saleem MA. The podocyte cytoskeleton–key to a functioning glomerulus in health and disease. Nature reviews. Nephrology. 2012;8:14–21. doi: 10.1038/nrneph.2011.151.
    1. Schwarz K, et al. Podocin, a raft-associated component of the glomerular slit diaphragm, interacts with CD2AP and nephrin. The Journal of clinical investigation. 2001;108:1621–1629. doi: 10.1172/JCI200112849.
    1. Vaidya VS, Ferguson MA, Bonventre JV. Biomarkers of acute kidney injury. Annual review of pharmacology and toxicology. 2008;48:463–493. doi: 10.1146/annurev.pharmtox.48.113006.094615.
    1. Beck LH, Jr., et al. M-type phospholipase A2 receptor as target antigen in idiopathic membranous nephropathy. The New England journal of medicine. 2009;361:11–21. doi: 10.1056/NEJMoa0810457.
    1. Wei C, et al. Circulating urokinase receptor as a cause of focal segmental glomerulosclerosis. Nature medicine. 2011;17:952–960. doi: 10.1038/nm.2411.
    1. Flint RS, et al. Probing the urinary proteome of severe acute pancreatitis. HPB: the official journal of the International Hepato Pancreato Biliary Association. 2007;9:447–455. doi: 10.1080/13651820701721744.
    1. Uhlen M, et al. Proteomics. Tissue-based map of the human proteome. Science. 2015;347:1260419. doi: 10.1126/science.1260419.
    1. Tran JC, et al. Mapping intact protein isoforms in discovery mode using top-down proteomics. Nature. 2011;480:254–258. doi: 10.1038/nature10575.
    1. Cargile BJ, Sevinsky JR, Essader AS, Eu JP, Stephenson JL., Jr. Calculation of the isoelectric point of tryptic peptides in the pH 3.5–4.5 range based on adjacent amino acid effects. Electrophoresis. 2008;29:2768–2778. doi: 10.1002/elps.200700701.
    1. Rabilloud T, et al. Power and limitations of electrophoretic separations in proteomics strategies. Mass spectrometry reviews. 2009;28:816–843. doi: 10.1002/mas.20204.
    1. Vaezzadeh AR, Briscoe AC, Steen H, Lee RS. One-step sample concentration, purification, and albumin depletion method for urinary proteomics. Journal of proteome research. 2010;9:6082–6089. doi: 10.1021/pr100924s.
    1. Guldbrandsen A, et al. In-depth characterization of the cerebrospinal fluid (CSF) proteome displayed through the CSF proteome resource (CSF-PR) Molecular & cellular proteomics: MCP. 2014;13:3152–3163. doi: 10.1074/mcp.M114.038554.
    1. Wisniewski JR, Zougman A, Nagaraj N, Mann M. Universal sample preparation method for proteome analysis. Nature methods. 2009;6:359–362. doi: 10.1038/nmeth.1322.
    1. Wang Y, et al. Reversed-phase chromatography with multiple fraction concatenation strategy for proteome profiling of human MCF10A cells. Proteomics. 2011;11:2019–2026. doi: 10.1002/pmic.201000722.
    1. Nesvizhskii AI, Keller A, Kolker E, Aebersold R. A statistical model for identifying proteins by tandem mass spectrometry. Analytical chemistry. 2003;75:4646–4658. doi: 10.1021/ac0341261.
    1. Schwanhausser B, et al. Global quantification of mammalian gene expression control. Nature. 2011;473:337–342. doi: 10.1038/nature10098.
    1. Hauck SM, et al. Deciphering membrane-associated molecular processes in target tissue of autoimmune uveitis by label-free quantitative mass spectrometry. Molecular & cellular proteomics: MCP. 2010;9:2292–2305. doi: 10.1074/mcp.M110.001073.
    1. Satoskar AA, et al. Characterization of glomerular diseases using proteomic analysis of laser capture microdissected glomeruli. Modern pathology: an official journal of the United States and Canadian Academy of Pathology, Inc. 2012;25:709–721. doi: 10.1038/modpathol.2011.205.
    1. Skoberne A, Konieczny A, Schiffer M. Glomerular epithelial cells in the urine: what has to be done to make them worthwhile? American journal of physiology. Renal physiology. 2009;296:F230–241. doi: 10.1152/ajprenal.90507.2008.
    1. Fukuda H, et al. Podocin is translocated to cytoplasm in puromycin aminonucleoside nephrosis rats and in poor-prognosis patients with IgA nephropathy. Cell and tissue research. 2015;360:391–400. doi: 10.1007/s00441-014-2100-9.
    1. Mansour H, et al. T-cell transcriptome analysis points up a thymic disorder in idiopathic nephrotic syndrome. Kidney international. 2005;67:2168–2177. doi: 10.1111/j.1523-1755.2005.00322.x.
    1. Kimura M, et al. Expression of alpha-actinin-4 in human diabetic nephropathy. Internal medicine. 2008;47:1099–1106. doi: 10.2169/internalmedicine.47.0352.
    1. Kaplan JM, et al. Mutations in ACTN4, encoding alpha-actinin-4, cause familial focal segmental glomerulosclerosis. Nature genetics. 2000;24:251–256. doi: 10.1038/73456.
    1. Kubiak-Wlekly A, et al. The Comparison of the Podocyte Expression of Synaptopodin, CR1 and Neprilysin in Human Glomerulonephritis: Could the Expression of CR1 be Clinically Relevant? International journal of biomedical science: IJBS. 2009;5:28–36.
    1. Johnstone DB, et al. Podocyte-specific deletion of Myh9 encoding nonmuscle myosin heavy chain 2A predisposes mice to glomerulopathy. Molecular and cellular biology. 2011;31:2162–2170. doi: 10.1128/MCB.05234-11.
    1. Yard, B. A. et al. Decreased glomerular expression of agrin in diabetic nephropathy and podocytes, cultured in high glucose medium. Experimental nephrology9, 214–222, doi:52614 (2001).
    1. Joosten SA, et al. Antibody response against the glomerular basement membrane protein agrin in patients with transplant glomerulopathy. American journal of transplantation: official journal of the American Society of Transplantation and the American Society of Transplant Surgeons. 2005;5:383–393. doi: 10.1111/j.1600-6143.2005.00690.x.
    1. Kashtan CE, Kim Y. Distribution of the alpha 1 and alpha 2 chains of collagen IV and of collagens V and VI in Alport syndrome. Kidney international. 1992;42:115–126. doi: 10.1038/ki.1992.269.
    1. Kim Y, et al. Differential expression of basement membrane collagen chains in diabetic nephropathy. The American journal of pathology. 1991;138:413–420.
    1. Kim Y, et al. Differential expression of basement membrane collagen in membranous nephropathy. The American journal of pathology. 1991;139:1381–1388.
    1. Setty S, et al. Differential expression of laminin isoforms in diabetic nephropathy and other renal diseases. Modern pathology: an official journal of the United States and Canadian Academy of Pathology, Inc. 2012;25:859–868. doi: 10.1038/modpathol.2011.216.
    1. Jim B, et al. Dysregulated nephrin in diabetic nephropathy of type 2 diabetes: a cross sectional study. PloS one. 2012;7:e36041. doi: 10.1371/journal.pone.0036041.
    1. Kim JM, et al. CD2-associated protein haploinsufficiency is linked to glomerular disease susceptibility. Science. 2003;300:1298–1300. doi: 10.1126/science.1081068.
    1. Ye H, et al. Urinary podocalyxin positive-element occurs in the early stage of diabetic nephropathy and is correlated with a clinical diagnosis of diabetic nephropathy. Journal of diabetes and its complications. 2014;28:96–100. doi: 10.1016/j.jdiacomp.2013.08.006.
    1. Tufro A, Veron D. VEGF and podocytes in diabetic nephropathy. Seminars in nephrology. 2012;32:385–393. doi: 10.1016/j.semnephrol.2012.06.010.
    1. El-Koraie AF, Baddour NM, Adam AG, El-Kashef EH, El Nahas AM. Cytoskeletal protein expression and regenerative markers in schistosomal nephropathy. Nephrology, dialysis, transplantation: official publication of the European Dialysis and Transplant Association - European Renal Association. 2002;17:803–812. doi: 10.1093/ndt/17.5.803.
    1. Birmingham DJ, et al. Relationship of Circulating Anti-C3b and Anti-C1q IgG to Lupus Nephritis and Its Flare. Clinical journal of the American Society of Nephrology: CJASN. 2016;11:47–53. doi: 10.2215/CJN.03990415.
    1. Ilhan N, Kahraman N, Seckin D, Ilhan N, Colak R. Apo E gene polymorphism on development of diabetic nephropathy. Cell biochemistry and function. 2007;25:527–532. doi: 10.1002/cbf.1348.
    1. Fugmann T, et al. Proteomic identification of vanin-1 as a marker of kidney damage in a rat model of type 1 diabetic nephropathy. Kidney international. 2011;80:272–281. doi: 10.1038/ki.2011.116.
    1. Wang QZ, et al. Cofilin1 is involved in hypertension-induced renal damage via the regulation of NF-kappaB in renal tubular epithelial cells. Journal of translational medicine. 2015;13:323. doi: 10.1186/s12967-015-0685-8.
    1. Castelletti F, et al. Mutations in FN1 cause glomerulopathy with fibronectin deposits. Proceedings of the National Academy of Sciences of the United States of America. 2008;105:2538–2543. doi: 10.1073/pnas.0707730105.
    1. Hanamura K, et al. Detection of myeloperoxidase in membranous nephropathy-like deposits in patients with anti-neutrophil cytoplasmic antibody-associated glomerulonephritis. Human pathology. 2011;42:649–658. doi: 10.1016/j.humpath.2010.08.020.
    1. Bonventre JV, Vaidya VS, Schmouder R, Feig P, Dieterle F. Next-generation biomarkers for detecting kidney toxicity. Nature biotechnology. 2010;28:436–440. doi: 10.1038/nbt0510-436.
    1. Goligorsky MS, Addabbo F, O’Riordan E. Diagnostic potential of urine proteome: a broken mirror of renal diseases. Journal of the American Society of Nephrology: JASN. 2007;18:2233–2239. doi: 10.1681/ASN.2006121399.
    1. Vaidya VS, Bonventre JV. Mechanistic biomarkers for cytotoxic acute kidney injury. Expert opinion on drug metabolism & toxicology. 2006;2:697–713. doi: 10.1517/17425255.2.5.697.
    1. Trof RJ, Di Maggio F, Leemreis J, Groeneveld AB. Biomarkers of acute renal injury and renal failure. Shock. 2006;26:245–253. doi: 10.1097/01.shk.0000225415.5969694.ce.
    1. Gal-Yam EN, Saito Y, Egger G, Jones PA. Cancer epigenetics: modifications, screening, and therapy. Annual review of medicine. 2008;59:267–280. doi: 10.1146/annurev.med.59.061606.095816.
    1. Thrailkill KM, et al. Microalbuminuria in type 1 diabetes is associated with enhanced excretion of the endocytic multiligand receptors megalin and cubilin. Diabetes care. 2009;32:1266–1268. doi: 10.2337/dc09-0112.
    1. Schaub JA, et al. Perioperative heart-type fatty acid binding protein is associated with acute kidney injury after cardiac surgery. Kidney international. 2015;88:576–583. doi: 10.1038/ki.2015.104.
    1. Wu I, Parikh CR. Screening for kidney diseases: older measures versus novel biomarkers. Clinical journal of the American Society of Nephrology: CJASN. 2008;3:1895–1901. doi: 10.2215/CJN.02030408.
    1. Ricci Z, Ronco C. Year in review: Critical Care 2004 - nephrology. Critical care. 2005;9:523–527. doi: 10.1186/cc3791.
    1. Iida T, et al. Decreased urinary calbindin 1 levels in proteinuric rats and humans with distal nephron segment injuries. Clinical and experimental nephrology. 2014;18:432–443. doi: 10.1007/s10157-013-0835-3.
    1. Sawai K, et al. Expression of CCN1 (CYR61) in developing, normal, and diseased human kidney. American journal of physiology. Renal physiology. 2007;293:F1363–1372. doi: 10.1152/ajprenal.00205.2007.
    1. Lameire N, Van Biesen W, Vanholder R. Acute renal failure. Lancet. 2005;365:417–430. doi: 10.1016/S0140-6736(05)70238-5.
    1. Nuyts GD, et al. Human urinary intestinal alkaline phosphatase as an indicator of S3-segment-specific alterations in incipient diabetic nephropathy. Nephrology, dialysis, transplantation: official publication of the European Dialysis and Transplant Association - European Renal Association. 1994;9:377–381.
    1. Adiyanti SS, Loho T. Acute Kidney Injury (AKI) biomarker. Acta medica Indonesiana. 2012;44:246–255.
    1. Ramesh G, Kwon O, Ahn K. Netrin-1: a novel universal biomarker of human kidney injury. Transplantation proceedings. 2010;42:1519–1522. doi: 10.1016/j.transproceed.2009.11.040.
    1. Mishra J, et al. Neutrophil gelatinase-associated lipocalin (NGAL) as a biomarker for acute renal injury after cardiac surgery. Lancet. 2005;365:1231–1238. doi: 10.1016/S0140-6736(05)74811-X.
    1. Yu XQ, et al. Osteopontin expression in progressive renal injury in remnant kidney: role of angiotensin II. Kidney international. 2000;58:1469–1480. doi: 10.1046/j.1523-1755.2000.00309.x.
    1. Liu Y, et al. Urinary interleukin 18 for detection of acute kidney injury: a meta-analysis. American journal of kidney diseases: the official journal of the National Kidney Foundation. 2013;62:1058–1067. doi: 10.1053/j.ajkd.2013.05.014.
    1. Varghese, S. A. et al. Identification of diagnostic urinary biomarkers for acute kidney injury. Journal of investigative medicine: the official publication of the American Federation for Clinical Research58, 612–620, doi:10.231/JIM.0b013e3181d473e7 (2010).

Source: PubMed

3
Subscribe