Serum uromodulin-a marker of kidney function and renal parenchymal integrity

Jürgen E Scherberich, Rudolf Gruber, Wolfgang Andreas Nockher, Erik Ilsø Christensen, Hans Schmitt, Victor Herbst, Matthias Block, Jürgen Kaden, Wolfgang Schlumberger, Jürgen E Scherberich, Rudolf Gruber, Wolfgang Andreas Nockher, Erik Ilsø Christensen, Hans Schmitt, Victor Herbst, Matthias Block, Jürgen Kaden, Wolfgang Schlumberger

Abstract

Background: An ELISA to analyse uromodulin in human serum (sUmod) was developed, validated and tested for clinical applications.

Methods: We assessed sUmod, a very stable antigen, in controls, patients with chronic kidney disease (CKD) stages 1-5, persons with autoimmune kidney diseases and recipients of a renal allograft by ELISA.

Results: Median sUmod in 190 blood donors was 207 ng/mL (women: men, median 230 versus 188 ng/mL, P = 0.006). sUmod levels in 443 children were 193 ng/mL (median). sUmod was correlated with cystatin C (rs = -0.862), creatinine (rs = -0.802), blood urea nitrogen (BUN) (rs = -0.645) and estimated glomerular filtration rate (eGFR)-cystatin C (rs = 0.862). sUmod was lower in systemic lupus erythematosus-nephritis (median 101 ng/mL), phospholipase-A2 receptor- positive glomerulonephritis (median 83 ng/mL) and anti-glomerular basement membrane positive pulmorenal syndromes (median 37 ng/mL). Declining sUmod concentrations paralleled the loss of kidney function in 165 patients with CKD stages 1-5 with prominent changes in sUmod within the 'creatinine blind range' (71-106 µmol/L). Receiver-operating characteristic analysis between non-CKD and CKD-1 was superior for sUmod (AUC 0.90) compared with eGFR (AUC 0.39), cystatin C (AUC 0.39) and creatinine (AUC 0.27). sUmod rapidly recovered from 0 to 62 ng/mL (median) after renal transplantation in cases with immediate graft function and remained low in delayed graft function (21 ng/mL, median; day 5-9: relative risk 1.5-2.9, odds ratio 1.5-6.4). Immunogold labelling disclosed that Umod is transferred within cytoplasmic vesicles to both the apical and basolateral plasma membrane. Umod revealed a disturbed intracellular location in kidney injury.

Conclusions: We conclude that sUmod is a novel sensitive kidney-specific biomarker linked to the structural integrity of the distal nephron and to renal function.

Keywords: kidney failure; renal biomarker; serum assay; thick ascending limb; uromodulin.

© The Author 2017. Published by Oxford University Press on behalf of ERA-EDTA.

Figures

FIGURE 1
FIGURE 1
Relationship between serum uromodulin and creatinine (A), blood urea nitrogen (B), cystatin C (C) and eGFRcystC (D). rs: Spearman’s rank correlation coefficient.
FIGURE 2
FIGURE 2
Serum uromodulin concentration of patients without kidney diseases (non-CKD) and patients with CKD (stage 1 to stage 5) based on eGFRcysC assignment.
FIGURE 3
FIGURE 3
ROC analysing the ability of different parameters to differentiate patients without kidney disease (non-CKD) and CKD stage 1; BUN =  blood urea nitrogen, eGFR = estimated glomerular filtration rate, AUC =  area under the curve.
FIGURE 4
FIGURE 4
Post-kidney transplant comparison of serum uromodulin and creatinine in patients with immediate and delayed graft function.
FIGURE 5
FIGURE 5
Immunohistochemical distribution of Umod in the normal human kidney (ac); semithin cryosections. (a) Apical, basolateral and cytoplasmatic staining, (b) transition of the thin ascending limb (negative) to the TAL (positive, arrows), (c) macula densa reveals a faint staining pattern only (arrowheads).
FIGURE 6
FIGURE 6
Immunogold labelling of Umod revealing the transport of the antigen within cytoplasmatic vesicles (small arrowheads) to the apical (a) as well as to the basolateral cell pole (b). Shuttle of uromodulin (c) from the endoplasmatic reticulum (RER) to the Golgi apparatus (G) (inset to C), through cytoplasmic vesicles (arrowheads). Basolateral membrane (BM) labelling (arrow). M, mitochondria.
FIGURE 7
FIGURE 7
Cartoon demonstrating the proposed bidirectional physiological transfer of Umod to the urinary space and to the tubulointerstitial space. As shown in Figure 6b, Umod is physiologically released from basolateral plasma membranes and membrane infoldings into the interstitium and enters the blood compartment (a). Umod serum levels are correlated with eGFR. sUmod concentrations may depend on the cellular integrity of the TAL segment. Therefore, sUmod might be released into serum at a decreased rate in renal injury (b).

References

    1. Tamm I, Horsfall FL.. Characterization and separation of an inhibitor of viral hemagglutination present in urine. Proc Soc Exp Biol Med 1990; 74: 106–108
    1. Devuyst O, Dahan K, Pirson Y.. Tamm–Horsfall protein or uromodulin: new ideas about an old molecule. Nephrol Dial Transplant 2005; 20: 1290–1294
    1. Rampoldi L, Scolari F, Amoroso A. et al. The rediscovery of uromodulin (Tamm–Horsfall protein): from tubulointerstitial nephropathy to chronic kidney disease. Kidney Int 2011; 80: 338–347
    1. Mount DB. Thick ascending limb of the loop of Henle. Clin J Am Soc Nephrol 2014; 9: 1974–1986
    1. Cavallone D, Malagolini N, Serafini-Cessi F.. Mechanism of release of urinary Tamm–Horsfall glycoprotein from the kidney GPI-anchored counterpart. Biochem Biophys Res Commun 2001; 280: 110–114
    1. Pak J, Pu X, Zhang ZT. et al. Tamm–Horsfall protein binds to type 1 fimbriated Escherichia coli and prevent E. coli from binding to uroplakin Ia and Ib receptors. J Biol Chem 2001; 276: 9924–9930
    1. Raffi HS, Bates JM Jr, Laszik Z. et al. Tamm–Horsfall protein protects against urinary tract infection by Proteus mirabilis. J Urol 2009; 181: 2332–2338
    1. Serafini-Cessa F, Monti A, Cavallone D.. N-glycans carried by Tamm–Horsfall glycoprotein have a crucial role in the defense against urinary tract diseases. Glycoconj J 2005; 22: 383–394
    1. Mo L, Huang HY, Zhu XH. et al. Tamm–Horsfall protein is a critical renal defense factor protecting against calcium oxalate crystal formation. Kidney Int 2004; 66: 1159–1166
    1. Graham LA, Padmanabhan S, Fraser NJ. et al. Validation of Uromodulin as a candidate gene for human essential hypertension. Hypertension 2014; 63: 551–558
    1. Zacchia M, Capasso G.. The importance of uromodulin as regulator of salt reabsorption along the thick ascending limb. Nephrol Dial Transplant 2015; 30: 158–160
    1. Trudu M, Janas S, Lanzani C. et al. Common noncoding UMOD gene variants induce salt-sensitive hypertension and kidney damage by increasing uromodulin expression. Nat Med 2013; 19: 1655–1660
    1. Scolari F, Izzi C, Ghiggeri GM.. Uromodulin: from monogenic to multifactorial diseases. Nephrol Dial Transplant 2015; 30: 1250–1256
    1. Kreft B, Jabs WJ, Laskay T. et al. Polarized expression of Tamm–Horsfall protein by renal tubular epithelial cells activates human granulocytes. Infect Immun 2002; 70: 2650–2656
    1. Iorember FM, Vehaskari VM.. Uromodulin: old friend with new roles in health and disease. Pediatr Nephrol 2014; 29: 1151–1158
    1. Säemann MD, Weichhart T, Zeyda M. et al. Tamm–Horsfall glycoprotein links innate immune cell activation with adaptive immunity via a Toll-like receptor-4-dependent mechanism. J Clin Invest 2005; 115: 468–475
    1. Darisipudi MN, Thomasova D, Mulay SR. et al. Uromodulin triggers IL-1ß-dependent innate immunity via the NLRP3 inflammasome. J Am Soc Nephrol 2012; 23: 1783–1789
    1. Schley G, Klanke B, Schödel J. et al. Hypoxia-inducible transcription factors stabilization in the thick ascending limb protects against ischemic acute kidney injury. J Am Soc Nephrol 2011; 22: 2004–2015
    1. Brezis M, Rosen S.. Hypoxia of the renal medulla—its implications for disease. N Engl J Med 1995; 332: 647–655
    1. Heyman SN, Rosenberger C, Rosen S.. Experimental ischemia—reperfusion: biases and myths—the proximal vs. distal hypoxic tubular injury debate revisited. Kidney Int 2010; 77: 9–16
    1. Rosen S, Stillman IE.. Acute tubular necrosis is a syndrome of physiologic and pathologic dissociation. J Am Soc Nephrol 2008; 19: 871–875
    1. Srichai MB, Hao C, Davis L. et al. Apoptosis of the thick ascending limb results in acute kidney injury. J Am Soc Nephrol 2008; 19: 1538–1546
    1. Sharfuddin AA, Molitoris BA.. Pathophysiology of ischemic acute kidney injury. Nat Rev Nephrol 2011; 7: 189–200
    1. Thornley C, Dawnay A, Cattell WR.. Human Tamm–Horsfall glycoprotein: urinary and plasma levels in normal subjects and patients with renal disease determined by a fully validated radioimmunoassay. Clin Sci (Lond) 1985; 68: 529–535
    1. Prajczer S, Heidenreich U, Pfaller W. et al. Evidence for a role of uromodulin in chronic kidney disease progression. Nephrol Dial Transplant 2010; 25: 1896–1903
    1. Risch L, Lhotta K, Meier D. et al. The serum uromodulin level is associated with kidney function. Clin Chem Lab Med 2014; 52: 1755–1761
    1. El-Achkar TM, McCracken R, Liu Y. et al. Tamm–Horsfall protein translocates to the basolateral domain of thick ascending limbs, interstitium, and circulation during recovery from acute kidney injury. Am J Physiol Renal Physiol 2013; 304: F1066–F1075
    1. Kaden J, Groth J, May G. et al. Urinary Tamm–Horsfall protein as a marker of renal transplant function. Urol Res 1994; 22: 131–136
    1. Kobayashi K, Fukuoka S.. Conditions for solubilization of Tamm–Horsfall protein/uromodulin in human urine and establishment of a sensitive and accurate enzyme-linked immunosorbent assay (ELISA) method. Arch Biochem Biophys 2001; 388: 113–120
    1. Catalano C, Torffvit O.. Urinary excretion of Tamm–Horsfall protein in normotensive, normo-albuminuric type 1 diabetic patients. Nephron 1996; 72: 436–441
    1. Youhanna S, Weber J, Beaujean V. et al. Determination of uromodulin in human urine: influence of storage and processing. Nephrol Dial Transplant 2014; 29: 136–145
    1. Shlipak MG, Li Y, Fox C. et al. Uromodulin concentrations are not associated with incident CKD among persons with coronary artery disease. BMC Nephrol 2011; 12: 2.
    1. Zhou J, Chen Y, Liu Y. et al. Urinary uromodulin excretion predicts progression of chronic kidney disease resulting from IgA nephropathy. PLoS One 2013; 8: e71023.
    1. Garimella PS, Biggs ML, Katz R. et al. Urinary uromodulin, kidney function, and cardiovascular disease in elderly adults. Kidney Int 2015; 88: 1126–1134
    1. Köttgen A, Hwang SJ, Larson MG. et al. Uromodulin levels associate with common UMOD variant and risk for incident CKD. J Am Soc Nephrol 2010; 21: 337–344
    1. Olden M, Corre T, Hayward C. et al. Common variants in UMOD associate with urinary uromodulin levels: a meta-analysis. J Am Soc Nephrol 2014; 25: 1869–1882
    1. Kumar S. Mechanisms of injury in Uromodulin-associated kidney disease. J Am Soc Nephrol 2007; 18: 10–12
    1. Bleyer AJ, Hart PS, Kmoch S.. Hereditary interstitial kidney disease. Semin Nephrol 2010; 30: 366–373
    1. Ekici AB, Hackenbeck T, Morinière V. et al. Renal fibrosis is the common feature of autosomal dominant tubulointerstitial kidney diseases caused by mutations in mucin 1 or uromodulin. Kidney Int 2014; 86: 589–599
    1. Dahan K, Devuyst O, Smaers M. et al. A cluster of mutations in the UMOD gene causes familial juvenile hyperuricemic nephropathy with abnormal expression of uromodulin. J Am Soc Nephrol 2003; 14: 2883–2893
    1. Hart TC, Gorry MC, Hart PS. et al. Mutations of the UMOD gene are responsible for medullary cystic kidney disease 2 and familial juvenile hyperuricaemic nephropathy. J Med Genet 2002; 39: 882–892
    1. Reznichenko A, Böger CA, Snieder H. et al. UMOD as a susceptibility gene for end-stage renal disease. BMC Med Genet 2012; 13: 78.
    1. Pruijm M, Ponte B, Ackermann D. et al. Association of urinary Uromodulin with clinical characteristics and markers of tubular function in the general population. Clin J Am Soc Nephrol 2016; 11: 70–80
    1. Kaden J. Optimal management of induction therapy with ATG in kidney allograft rejection. Int J Immunother 1999; 15: 115–124
    1. Glauser A, Hochreiter W, Jaeger P. et al. Determinants of urinary excretion of Tamm–Horsfall protein in non-selected kidney stone formers and healthy subjects. Nephrol Dial Transplant 2000; 15: 1580–1587
    1. Trachtenberg F, Barregard L.. The effect of age, sex, and race on urinary markers of kidney damage in children. Am J Kidney Dis 2007; 50: 938–945
    1. Neugarten J, Golestaneh L.. Gender and the prevalence and progression of renal disease. Adv Chronic Kidney Dis 2013; 20: 390–395
    1. Silbiger SR. Raging hormones: gender and renal disease. Kidney Int 2011; 79: 382–384
    1. Wang X, Vrtiska TJ, Avula RT. et al. Age, kidney function, and risk factors associate differentially with cortical and medullary volumes of the kidney. Kidney Int 2014; 85: 677–685
    1. Steubl D, Block M, Herbst V. et al. Plasma uromodulin correlates with kidney function and identifies early stages in chronic kidney disease patients. Medicine 2016; 95: e3011.
    1. Nath KA. Tubulointerstitial changes as a major determinant in the progression of renal damage. Am J Kidney Dis 1992; 20: 1–17
    1. Healy E, Brady HR.. Role of tubule epithelial cells in the pathogenesis of tubulointerstitial fibrosis induced by glomerular disease. Curr Opin Nephrol Hypertens 1998; 7: 525–530
    1. Farris AB, Ellis CL, Rogers TE. et al. Renal medullary and cortical correlates in fibrosis, epithelial mass, microvascularity, and microanatomy using whole slide image analysis morphometry. PLoS One 11: e0161019
    1. El-Achkar TM, Wu XR.. Uromodulin in kidney injury: an instigator, bystander, or protector? Am J Kidney 2012; 59: 452–461
    1. Jennings P, Aydin S, Kotanko P. et al. Membrane targeting and secretion of mutant uromodulin in familial juvenile hyperuricemic nephropathy. J Am Soc Nephrol 2007; 18: 264–273
    1. Biesen van W, Nagler EV.. A Swiss army knife for estimating kidney function: why new equations will not solve the real problem. Nephrol Dial Transplant 2016; 31: 685–687
    1. Gill J, Dong J, Rose C. et al. The risk of allograft failure and the survival benefit of kidney transplantation are complicated by delayed graft function. Kidney Int 2016; 89: 1331–1336
    1. Scherberich JE. The king’s new cloths—the infinite history of kidney tissue proteinuria (histuria) as a renal biomarker. Nieren- und Hochdruckkrankh 2012; 41: 436–450
    1. Scherberich JE, Wolf G.. Disintegration and recovery of kidney membrane proteins: consequence of acute and chronic renal failure. Kidney Int Suppl 1994; 47: S52–S57
    1. Johnson KR. Strengths and weaknesses of renal markers as risk factors and surrogate markers. Kidney Int 2011; 79: 1272–1274
    1. Filler G, Huang SH, Lindsay RM.. The search for more reliable estimated GFR biomarkers. Am J Kidney Dis 2016; 67: 5–8

Source: PubMed

3
Suscribir