Abstract
Uromodulin (also known as Tamm–Horsfall protein) is a kidney-specific glycoprotein secreted bidirectionally into urine and into the circulation, and it is the most abundant protein in normal urine. Although the discovery of uromodulin predates modern medicine, its significance in health and disease has been rather enigmatic. Research studies have gradually revealed that uromodulin exists in multiple forms and has important roles in urinary and systemic homeostasis. Most uromodulin in urine is polymerized into highly organized filaments, whereas non-polymeric uromodulin is detected both in urine and in the circulation, and can have distinct roles. The interactions of uromodulin with the immune system, which were initially reported to be a key role of this protein, are now better understood. Moreover, the discovery that uromodulin is associated with a spectrum of kidney diseases, including acute kidney injury, chronic kidney disease and autosomal-dominant tubulointerstitial kidney disease, has further accelerated investigations into the role of this protein. These discoveries have prompted new questions and ushered in a new era in uromodulin research. Here, we delineate the latest discoveries in uromodulin biology and its emerging roles in modulating kidney and systemic diseases, and consider future directions, including its potential clinical applications.
Key points
-
Most urinary uromodulin undergoes polymerization into filaments and has important roles in maintaining urinary homeostasis. This polymerization occurs on the apical membrane and is highly organized.
-
Non-polymeric uromodulin, an increasingly recognized form, has distinct kidney and systemic roles. Recognizing the specific form and site of action is crucial to understanding the function of uromodulin.
-
Acute kidney injury is characterized by uromodulin deficiency. Non-polymerizing uromodulin supplementation might improve the course of acute kidney injury and prevent the transition to chronic kidney disease.
-
Single-nucleotide polymorphisms in UMOD and PDILT are strongly linked to the risk and progression of chronic kidney disease. Whether this effect depends on expression of a specific form of uromodulin remains to be clarified.
-
Retention and aggregation of mutant uromodulin in the endoplasmic reticulum causes autosomal-dominant tubulointerstitial kidney disease. Clearance of mutant uromodulin might be a promising therapeutic approach.
This is a preview of subscription content, access via your institution
Access options
Access Nature and 54 other Nature Portfolio journals
Get Nature+, our best-value online-access subscription
$29.99 / 30 days
cancel any time
Subscribe to this journal
Receive 12 print issues and online access
$209.00 per year
only $17.42 per issue
Buy this article
- Purchase on SpringerLink
- Instant access to full article PDF
Prices may be subject to local taxes which are calculated during checkout
Similar content being viewed by others
References
Devuyst, O., Olinger, E. & Rampoldi, L. Uromodulin: from physiology to rare and complex kidney disorders. Nat. Rev. Nephrol. 13, 525–544 (2017).
El-Achkar, T. M. & Wu, X.-R. Uromodulin in kidney injury: an instigator, bystander, or protector? Am. J. Kidney Dis. 59, 452–461 (2012).
Schaeffer, C., Devuyst, O. & Rampoldi, L. Uromodulin: roles in health and disease. Annu. Rev. Physiol. 83, 477–501 (2021).
Fairley, J. K., Owen, J. E. & Birch, D. F. Protein composition of urinary casts from healthy subjects and patients with glomerulonephritis. Br. Med. J. 287, 1838–1840 (1983).
Rovida, C. L. Conclusione degli studi intorno all’origine istologica dei cilindri dell’urina. Clin. Bologna 2a, 303–306 (1873).
Tamm, I. & Horsfall, F. L. Characterization and separation of an inhibitor of viral hemagglutination present in urine. Proc. Soc. Exp. Biol. Med. 74, 106–108 (1950).
Muchmore, A. V. & Decker, J. M. Uromodulin: a unique 85-kilodalton immunosuppressive glycoprotein isolated from urine of pregnant women. Science 229, 479–481 (1985).
Pennica, D. et al. Identification of human uromodulin as the Tamm-Horsfall urinary glycoprotein. Science 236, 83–88 (1987).
Micanovic, R., LaFavers, K., Garimella, P. S., Wu, X.-R. & El-Achkar, T. M. Uromodulin (Tamm–Horsfall protein): guardian of urinary and systemic homeostasis. Nephrol. Dial. Transpl. 35, 33–43 (2020).
Sikri, K. L., Foster, C. L., MacHugh, N. & Marshall, R. D. Localization of Tamm-Horsfall glycoprotein in the human kidney using immuno-fluorescence and immuno-electron microscopical techniques. J. Anat. 132, 597–605 (1981).
Tokonami, N. et al. Uromodulin is expressed in the distal convoluted tubule, where it is critical for regulation of the sodium chloride cotransporter NCC. Kidney Int. 94, 701–715 (2018).
Lee, J. W., Chou, C.-L. & Knepper, M. A. Deep sequencing in microdissected renal tubules identifies nephron segment-specific transcriptomes. J. Am. Soc. Nephrol. 26, 2669–2677 (2015).
Chen, L., Chou, C.-L. & Knepper, M. A. A comprehensive map of mRNAs and their isoforms across all 14 renal tubule segments of mouse. J. Am. Soc. Nephrol. 32, 897–912 (2021).
Uhlén, M. et al. Proteomics. Tissue-based map of the human proteome. Science 347, 1260419 (2015).
Bachmann, S., Koeppen-Hagemann, I. & Kriz, W. Ultrastructural localization of Tamm-Horsfall glycoprotein (THP) in rat kidney as revealed by protein A-gold immunocytochemistry. Histochemistry 83, 531–538 (1985).
Pook, M. A. et al. Localization of the Tamm-Horsfall glycoprotein (uromodulin) gene to chromosome 16p12.3-16p13.11. Ann. Hum. Genet. 57, 285–290 (1993).
Srivastava, R., Micanovic, R., El-Achkar, T. M. & Janga, S. C. An intricate network of conserved DNA upstream motifs and associated transcription factors regulate the expression of uromodulin gene. J. Urol. 192, 981–989 (2014).
Gresh, L. et al. A transcriptional network in polycystic kidney disease. EMBO J. 23, 1657–1668 (2004).
Trudu, M. et al. Common noncoding UMOD gene variants induce salt-sensitive hypertension and kidney damage by increasing uromodulin expression. Nat. Med. 19, 1655–1660 (2013). This study examined the functional significance of common UMOD variants using clinical data and experimental models.
Köttgen, A. et al. Uromodulin levels associate with a common UMOD variant and risk for incident CKD. J. Am. Soc. Nephrol. 21, 337–344 (2010).
Troyanov, S. et al. Clinical, genetic, and urinary factors associated with uromodulin excretion. Clin. J. Am. Soc. Nephrol. 11, 62–69 (2016).
Olden, M. et al. Common variants in UMOD associate with urinary uromodulin levels: a meta-analysis. J. Am. Soc. Nephrol. 25, 1869–1882 (2014).
Li, Y. et al. Genome-wide studies reveal factors associated with circulating uromodulin and its relationships to complex diseases. JCI Insight 7, e157035 (2022). A meta-analysis of GWAS of circulating uromodulin that identified several loci associated with circulating uromodulin levels.
Wuttke, M. et al. A catalog of genetic loci associated with kidney function from analyses of a million individuals. Nat. Genet. 51, 957–972 (2019).
Morris, A. P. et al. Trans-ethnic kidney function association study reveals putative causal genes and effects on kidney-specific disease aetiologies. Nat. Commun. 10, 29 (2019).
Nanamatsu, A., Micanovic, R., Khan, S., El-Achkar, T. M. & LaFavers, K. A. Healthy women have higher systemic uromodulin levels: identification of uromodulin as an estrogen responsive gene. Kidney360 4, e1302–e1307 (2023).
Ying, W. Z. & Sanders, P. W. Dietary salt regulates expression of Tamm-Horsfall glycoprotein in rats. Kidney Int. 54, 1150–1156 (1998).
Olinger, E. et al. Hepsin-mediated processing of uromodulin is crucial for salt-sensitivity and thick ascending limb homeostasis. Sci. Rep. 9, 12287 (2019).
Ponte, B. et al. Uromodulin, salt, and 24-hour blood pressure in the general population. Clin. J. Am. Soc. Nephrol. 16, 787–789 (2021).
LaFavers, K. A. et al. Water loading and uromodulin secretion in healthy individuals and idiopathic calcium stone formers. Clin. J. Am. Soc. Nephrol. 18, 1059–1067 (2023). Experimental study showing that water loading induces urinary uromodulin secretion in humans.
Torffvit, O., Melander, O. & Hultén, U. L. Urinary excretion rate of Tamm-Horsfall protein is related to salt intake in humans. Nephron Physiol. 97, p31–p36 (2004).
Mladinov, D., Liu, Y., Mattson, D. L. & Liang, M. MicroRNAs contribute to the maintenance of cell-type-specific physiological characteristics: miR-192 targets Na+/K+-ATPase β1. Nucleic Acids Res. 41, 1273–1283 (2013).
Resnick, J. S., Sisson, S. & Vernier, R. L. Tamm-Horsfall protein. Abnormal localization in renal disease. Lab. Invest. 38, 550–555 (1978).
McGiven, A. R., Hunt, J. S., Day, W. A. & Bailey, R. R. Tamm-Horsfall protein in the glomerular capsular space. J. Clin. Pathol. 31, 620–625 (1978).
Maydan, O. et al. Uromodulin deficiency alters tubular injury and interstitial inflammation but not fibrosis in experimental obstructive nephropathy. Physiol. Rep. 6, e13654 (2018).
Fasth, A. L., Hoyer, J. R. & Seiler, M. W. Extratubular Tamm-Horsfall protein deposits induced by ureteral obstruction in mice. Clin. Immunol. Immunopathol. 47, 47–61 (1988).
Sato, S. et al. Transcription factor Spi-B-dependent and -independent pathways for the development of Peyer’s patch M cells. Mucosal Immunol. 6, 838–846 (2013).
Yanagihara, S. et al. Uromodulin-SlpA binding dictates Lactobacillus acidophilus uptake by intestinal epithelial M cells. Int. Immunol. 29, 357–363 (2017).
Knauf, F., Brewer, J. R. & Flavell, R. A. Immunity, microbiota and kidney disease. Nat. Rev. Nephrol. 15, 263–274 (2019).
O’Donnell, J. A., Zheng, T., Meric, G. & Marques, F. Z. The gut microbiome and hypertension. Nat. Rev. Nephrol. 19, 153–167 (2023).
Evenepoel, P., Stenvinkel, P., Shanahan, C. & Pacifici, R. Inflammation and gut dysbiosis as drivers of CKD-MBD. Nat. Rev. Nephrol. 19, 646–657 (2023).
LaFavers, K. A. et al. The kidney protects against sepsis by producing systemic uromodulin. Am. J. Physiol. Renal Physiol. 323, F212–F226 (2022).
Fontan, E., Fauve, R. M., Hevin, B. & Jusforgues, H. Immunostimulatory mouse granuloma protein. Proc. Natl Acad. Sci. USA 80, 6395–6398 (1983).
Rindler, M. J., Naik, S. S., Li, N., Hoops, T. C. & Peraldi, M. N. Uromodulin (Tamm-Horsfall glycoprotein/uromucoid) is a phosphatidylinositol-linked membrane protein. J. Biol. Chem. 265, 20784–20789 (1990).
Stsiapanava, A. et al. Structure of the decoy module of human glycoprotein 2 and uromodulin and its interaction with bacterial adhesin FimH. Nat. Struct. Mol. Biol. 29, 190–193 (2022). Definition and characterization of the D10C domain and decoy module of uromodulin by combining AlphaFold2 predictions, X-ray crystallography and Cryo-electron microscopy.
Yang, H., Wu, C., Zhao, S. & Guo, J. Identification and characterization of D8C, a novel domain present in liver-specific LZP, uromodulin and glycoprotein 2, mutated in familial juvenile hyperuricaemic nephropathy. FEBS Lett. 578, 236–238 (2004).
Schaeffer, C., Santambrogio, S., Perucca, S., Casari, G. & Rampoldi, L. Analysis of uromodulin polymerization provides new insights into the mechanisms regulating ZP domain-mediated protein assembly. Mol. Biol. Cell 20, 589–599 (2009).
Stsiapanava, A. et al. Cryo-EM structure of native human uromodulin, a zona pellucida module polymer. EMBO J. 39, e106807 (2020).
van Rooijen, J. J., Voskamp, A. F., Kamerling, J. P. & Vliegenthart, J. F. Glycosylation sites and site-specific glycosylation in human Tamm-Horsfall glycoprotein. Glycobiology 9, 21–30 (1999).
Weiss, G. L. et al. Architecture and function of human uromodulin filaments in urinary tract infections. Science 369, 1005–1010 (2020). Demonstration of the structure and function of urinary uromodulin filaments using cryo-electron tomography.
Easton, R. L., Patankar, M. S., Clark, G. F., Morris, H. R. & Dell, A. Pregnancy-associated changes in the glycosylation of Tamm-Horsfall glycoprotein. Expression of sialyl Lewis(x) sequences on core 2 type O-glycans derived from uromodulin. J. Biol. Chem. 275, 21928–21938 (2000).
Stoops, E. H. & Caplan, M. J. Trafficking to the apical and basolateral membranes in polarized epithelial cells. J. Am. Soc. Nephrol. 25, 1375–1386 (2014).
Tokonami, N., Olinger, E., Debaix, H., Houillier, P. & Devuyst, O. The excretion of uromodulin is modulated by the calcium-sensing receptor. Kidney Int. 94, 882–886 (2018).
Joseph, C. B. et al. Meta-GWAS reveals novel genetic variants associated with urinary excretion of uromodulin. J. Am. Soc. Nephrol. 33, 511–529 (2022).
Otterpohl, K. L. et al. Conditional Myh9 and Myh10 inactivation in adult mouse renal epithelium results in progressive kidney disease. JCI Insight 5, e138530 (2020).
Mariniello, M. et al. Uromodulin processing in DNAJB11-kidney disease. Kidney Int. S0085-2538, 00785–00788 (2023).
Schiano, G. et al. Allelic effects on uromodulin aggregates drive autosomal dominant tubulointerstitial kidney disease. EMBO Mol. Med. 15, e18242 (2023).
Brunati, M. et al. The serine protease hepsin mediates urinary secretion and polymerisation of Zona Pellucida domain protein uromodulin. Elife 4, e08887 (2015). Identification of hepsin as a crucial protease for releasing polymerizing uromodulin into urine.
Bokhove, M. et al. A structured interdomain linker directs self-polymerization of human uromodulin. Proc. Natl Acad. Sci. USA 113, 1552–1557 (2016).
Stanisich, J. J. et al. The cryo-EM structure of the human uromodulin filament core reveals a unique assembly mechanism. Elife 9, e60265 (2020).
Nanamatsu, A. et al. Vasopressin induces urinary uromodulin secretion by activating PKA (Protein Kinase A). Hypertension 77, 1953–1963 (2021).
Ecelbarger, C. A. et al. Localization and regulation of the rat renal Na+-K+-2Cl- cotransporter, BSC-1. Am. J. Physiol. 271, F619–F628 (1996).
Bachmann, S., Dawnay, A. B., Bouby, N. & Bankir, L. Tamm-Horsfall protein excretion during chronic alterations in urinary concentration and protein intake in the rat. Ren. Physiol. Biochem. 14, 236–245 (1991).
Schiano, G. et al. The urinary excretion of uromodulin is regulated by the potassium channel ROMK. Sci. Rep. 9, 19517 (2019).
Micanovic, R. et al. The kidney releases a nonpolymerizing form of uromodulin in the urine and circulation that retains the external hydrophobic patch domain. Am. J. Physiol. Renal Physiol. 322, F403–F418 (2022).
Mary, S. et al. Polymerization-incompetent uromodulin in the pregnant stroke-prone spontaneously hypertensive rat. Hypertension 69, 910–918 (2017).
Jovine, L., Qi, H., Williams, Z., Litscher, E. & Wassarman, P. M. The ZP domain is a conserved module for polymerization of extracellular proteins. Nat. Cell Biol. 4, 457–461 (2002).
Van, J. A. D. et al. Peptidomic analysis of urine from youths with early type 1 diabetes reveals novel bioactivity of uromodulin peptides in vitro. Mol. Cell Proteom. 19, 501–517 (2020).
El-Achkar, T. M. 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. 304, F1066–F1075 (2013).
Micanovic, R. et al. Tamm-Horsfall protein regulates mononuclear phagocytes in the kidney. J. Am. Soc. Nephrol. 29, 841–856 (2018).
Bates, J. M. et al. Tamm-Horsfall protein knockout mice are more prone to urinary tract infection: rapid communication. Kidney Int. 65, 791–797 (2004).
Mo, L. et al. Ablation of the Tamm-Horsfall protein gene increases susceptibility of mice to bladder colonization by type 1-fimbriated Escherichia coli. Am. J. Physiol. Renal Physiol. 286, F795–F802 (2004).
Coady, A., Ramos, A. R., Olson, J., Nizet, V. & Patras, K. A. Tamm-Horsfall protein protects the urinary tract against Candida albicans. Infect. Immun. 86, e00451–18 (2018).
Raffi, H. S., Bates, J. M., Laszik, Z. & Kumar, S. Tamm-Horsfall protein acts as a general host-defense factor against bacterial cystitis. Am. J. Nephrol. 25, 570–578 (2005).
Raffi, H. S., Bates, J. M., Laszik, Z. & Kumar, S. Tamm-Horsfall protein protects against urinary tract infection by Proteus mirabilis. J. Urol. 181, 2332–2338 (2009).
Bjugn, R. & Flood, P. R. Scanning electron microscopy of human urine and purified Tamm-Horsfall’s glycoprotein. Scand. J. Urol. Nephrol. 22, 313–315 (1988).
Pak, J., Pu, Y., Zhang, Z. T., Hasty, D. L. & Wu, X. R. Tamm-Horsfall protein binds to type 1 fimbriated Escherichia coli and prevents E. coli from binding to uroplakin Ia and Ib receptors. J. Biol. Chem. 276, 9924–9930 (2001).
Garimella, P. S. et al. Urinary uromodulin and risk of urinary tract infections: the Cardiovascular Health Study. Am. J. Kidney Dis. 69, 744–751 (2017).
Ghirotto, S. et al. The uromodulin gene locus shows evidence of pathogen adaptation through human evolution. J. Am. Soc. Nephrol. 27, 2983–2996 (2016).
Chen, W. C., Lin, H. S., Chen, H. Y., Shih, C. H. & Li, C. W. Effects of Tamm-Horsfall protein and albumin on calcium oxalate crystallization and importance of sialic acids. Mol. Urol. 5, 1–5 (2001).
Hallson, P. C., Choong, S. K., Kasidas, G. P. & Samuell, C. T. Effects of Tamm-Horsfall protein with normal and reduced sialic acid content upon the crystallization of calcium phosphate and calcium oxalate in human urine. Br. J. Urol. 80, 533–538 (1997).
Serafini-Cessi, 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. 22, 383–394 (2005).
Liu, Y. et al. Progressive renal papillary calcification and ureteral stone formation in mice deficient for Tamm-Horsfall protein. Am. J. Physiol. Renal Physiol. 299, F469–F478 (2010).
Mo, L. et al. Tamm-Horsfall protein is a critical renal defense factor protecting against calcium oxalate crystal formation. Kidney Int. 66, 1159–1166 (2004).
Mo, L. et al. Renal calcinosis and stone formation in mice lacking osteopontin, Tamm-Horsfall protein, or both. Am. J. Physiol. Renal Physiol. 293, F1935–F1943 (2007).
Wolf, M. T. F., Wu, X.-R. & Huang, C.-L. Uromodulin upregulates TRPV5 by impairing caveolin-mediated endocytosis. Kidney Int. 84, 130–137 (2013).
Gudbjartsson, D. F. et al. Association of variants at UMOD with chronic kidney disease and kidney stones-role of age and comorbid diseases. PLoS Genet. 6, e1001039 (2010).
Nie, M. et al. Uromodulin regulates renal magnesium homeostasis through the ion channel transient receptor potential melastatin 6 (TRPM6). J. Biol. Chem. 293, 16488–16502 (2018).
Fontan, E. et al. A 92-kDa human immunostimulatory protein. Proc. Natl Acad. Sci. USA 91, 8353–8357 (1994).
Fontan, E., Jusforgues-Saklani, H., Briend, E. & Fauve, R. M. Purification of a 92 kDa human immunostimulating glycoprotein obtained from the Tamm-Horsfall glycoprotein. J. Immunol. Methods 187, 81–84 (1995).
LaFavers, K. A. et al. Circulating uromodulin inhibits systemic oxidative stress by inactivating the TRPM2 channel. Sci. Transl. Med. 11, eaaw3639 (2019). Demonstration that circulating uromodulin suppresses systemic oxidative stress.
Alesutan, I. et al. Circulating uromodulin inhibits vascular calcification by interfering with pro-inflammatory cytokine signalling. Cardiovasc. Res. 117, 930–941 (2021).
Mutig, K. et al. Activation of the bumetanide-sensitive Na+,K+,2Cl− cotransporter (NKCC2) is facilitated by Tamm-Horsfall protein in a chloride-sensitive manner. J. Biol. Chem. 286, 30200–30210 (2011).
Renigunta, A. et al. Tamm-Horsfall glycoprotein interacts with renal outer medullary potassium channel ROMK2 and regulates its function. J. Biol. Chem. 286, 2224–2235 (2011).
Graham, L. A. et al. Validation of uromodulin as a candidate gene for human essential hypertension. Hypertension 63, 551–558 (2014).
Thomas, D. B., Davies, M., Peters, J. R. & Williams, J. D. Tamm Horsfall protein binds to a single class of carbohydrate specific receptors on human neutrophils. Kidney Int. 44, 423–429 (1993).
Patras, K. A. et al. Tamm–Horsfall glycoprotein engages human Siglec-9 to modulate neutrophil activation in the urinary tract. Immunol. Cell Biol. 95, 960–965 (2017).
El-Achkar, T. M. et al. Tamm-Horsfall protein-deficient thick ascending limbs promote injury to neighboring S3 segments in an MIP-2-dependent mechanism. Am. J. Physiol. Renal Physiol. 300, F999–F1007 (2011).
Micanovic, R. et al. Tamm-Horsfall protein regulates granulopoiesis and systemic neutrophil homeostasis. J. Am. Soc. Nephrol. 26, 2172–2182 (2015).
Zager, R. A., Cotran, R. S. & Hoyer, J. R. Pathologic localization of Tamm-Horsfall protein in interstitial deposits in renal disease. Lab. Invest. 38, 52–57 (1978).
Immler, R. et al. Extratubular polymerized uromodulin induces leukocyte recruitment and inflammation in vivo. Front. Immunol. 11, 588245 (2020).
Säemann, M. D. et al. Tamm-Horsfall glycoprotein links innate immune cell activation with adaptive immunity via a Toll-like receptor-4-dependent mechanism. J. Clin. Invest. 115, 468–475 (2005).
Pfistershammer, K. et al. Identification of the scavenger receptors SREC-I, Cla-1 (SR-BI), and SR-AI as cellular receptors for Tamm-Horsfall protein. J. Leukoc. Biol. 83, 131–138 (2008).
Darisipudi, M. N. et al. Uromodulin triggers IL-1β-dependent innate immunity via the NLRP3 inflammasome. J. Am. Soc. Nephrol. 23, 1783–1789 (2012).
Fontan, E., Saklani-Jusforgues, H. & Fauve, R. M. Immunostimulatory human urinary protein. Proc. Natl Acad. Sci. USA 89, 4358–4362 (1992).
McMahon, G. M. & Waikar, S. S. Biomarkers in nephrology: core Curriculum 2013. Am. J. Kidney Dis. 62, 165–178 (2013).
Scherberich, J. E. et al. Serum uromodulin-a marker of kidney function and renal parenchymal integrity. Nephrol. Dial. Transpl. 33, 284–295 (2018).
Youhanna, S. et al. Determination of uromodulin in human urine: influence of storage and processing. Nephrol. Dial. Transpl. 29, 136–145 (2014).
LaFavers, K. A., Micanovic, R., Sabo, A. R., Maghak, L. A. & El-Achkar, T. M. Evolving concepts in uromodulin biology, physiology, and its role in disease: a tale of two forms. Hypertension 79, 2409–2418 (2022).
Pivin, E. et al. Uromodulin and nephron mass. Clin. J. Am. Soc. Nephrol. 13, 1556–1557 (2018).
Lynn, K. L. & Marshall, R. D. Excretion of Tamm-Horsfall glycoprotein in renal disease. Clin. Nephrol. 22, 253–257 (1984).
Pruijm, M. et al. Associations of urinary uromodulin with clinical characteristics and markers of tubular function in the general population. Clin. J. Am. Soc. Nephrol. 11, 70–80 (2016).
Dawnay, A. B. & Cattell, W. R. Serum Tamm-Horsfall glycoprotein levels in health and in renal disease. Clin. Nephrol. 15, 5–8 (1981).
Risch, L. et al. The serum uromodulin level is associated with kidney function. Clin. Chem. Lab. Med. 52, 1755–1761 (2014).
Fedak, D. et al. Serum uromodulin concentrations correlate with glomerular filtration rate in patients with chronic kidney disease. Pol. Arch. Med. Wewn. 126, 995–1004 (2016).
Enko, D. et al. Individual uromodulin serum concentration is independent of glomerular filtration rate in healthy kidney donors. Clin. Chem. Lab. Med. 59, 563–570 (2021).
Then, C. et al. Serum uromodulin and risk for cardiovascular morbidity and mortality in the community-based KORA F4 study. Atherosclerosis 297, 1–7 (2020).
Denic, A. et al. Single-nephron glomerular filtration rate in healthy adults. N. Engl. J. Med. 376, 2349–2357 (2017).
Klein, S. L. & Flanagan, K. L. Sex differences in immune responses. Nat. Rev. Immunol. 16, 626–638 (2016).
Schiel, R. et al. Serum uromodulin in children and adolescents with type 1 diabetes mellitus and controls: its potential role in kidney health. Exp. Clin. Endocrinol. Diabetes 131, 142–152 (2023).
Leiherer, A. et al. Serum uromodulin is associated with impaired glucose metabolism. Medicine 96, e5798 (2017).
Then, C. et al. Serum uromodulin is associated with but does not predict type 2 diabetes in elderly KORA F4/FF4 study participants. J. Clin. Endocrinol. Metab. 104, 3795–3802 (2019).
Wiromrat, P. et al. Serum uromodulin is associated with urinary albumin excretion in adolescents with type 1 diabetes. J. Diabetes Complications 33, 648–650 (2019).
Bjornstad, P. et al. Serum uromodulin predicts less coronary artery calcification and diabetic kidney disease over 12 years in adults with type 1 diabetes: the CACTI study. Diabetes Care 42, 297–302 (2019).
Then, C. et al. Serum uromodulin is inversely associated with the metabolic syndrome in the KORA F4 study. Endocr. Connect. 8, 1363–1371 (2019).
Then, C. et al. Association of serum uromodulin with adipokines in dependence of type 2 diabetes. Cytokine 150, 155786 (2022).
Then, C. et al. Serum uromodulin is inversely associated with biomarkers of subclinical inflammation in the population-based KORA F4 study. Clin. Kidney J. 14, 1618–1625 (2021).
Hotamisligil, G. S. Inflammation and metabolic disorders. Nature 444, 860–867 (2006).
Bächle, H. et al. Uromodulin and its association with urinary metabolites: the German Chronic Kidney Disease Study. Nephrol. Dial. Transpl. 38, 70–79 (2023).
Patidar, K. R. et al. Admission plasma uromodulin and the risk of acute kidney injury in hospitalized patients with cirrhosis: a pilot study. Am. J. Physiol. Gastrointest. Liver Physiol. 317, G447–G452 (2019).
Garimella, P. S. et al. Association of preoperative urinary uromodulin with AKI after cardiac surgery. Clin. J. Am. Soc. Nephrol. 12, 10–18 (2017).
Bullen, A. L. et al. The SPRINT trial suggests that markers of tubule cell function in the urine associate with risk of subsequent acute kidney injury while injury markers elevate after the injury. Kidney Int. 96, 470–479 (2019).
Bennett, M. R., Pyles, O., Ma, Q. & Devarajan, P. Preoperative levels of urinary uromodulin predict acute kidney injury after pediatric cardiopulmonary bypass surgery. Pediatr. Nephrol. 33, 521–526 (2018).
Wen, Y. et al. Longitudinal biomarkers and kidney disease progression after acute kidney injury. JCI Insight 8, e167731 (2023).
Melchinger, H. et al. Urine uromodulin as a biomarker of kidney tubulointerstitial fibrosis. Clin. J. Am. Soc. Nephrol. 17, 1284–1292 (2022).
Puthumana, J. et al. Biomarkers of inflammation and repair in kidney disease progression. J. Clin. Invest. 131, e139927 (2021). Demonstration of the link between urinary uromodulin levels and kidney disease progression in clinical and experimental analyses.
Vasquez-Rios, G. et al. Joint modeling of clinical and biomarker data in acute kidney injury defines unique subphenotypes with differing outcomes. Clin. J. Am. Soc. Nephrol. 18, 716–726 (2023).
Steubl, D. et al. Association of serum uromodulin with ESKD and kidney function decline in the elderly: the Cardiovascular Health Study. Am. J. Kidney Dis. 74, 501–509 (2019).
Garimella, P. S. et al. Association of urinary uromodulin with kidney function decline and mortality: the health ABC study. Clin. Nephrol. 87, 278–286 (2017).
Leiherer, A. et al. The value of uromodulin as a new serum marker to predict decline in renal function. J. Hypertens. 36, 110–118 (2018).
Steubl, D. et al. Association of serum uromodulin with death, cardiovascular events, and kidney failure in CKD. Clin. J. Am. Soc. Nephrol. 15, 616–624 (2020).
Garimella, P. S. et al. Urinary uromodulin, kidney function, and cardiovascular disease in elderly adults. Kidney Int. 88, 1126–1134 (2015).
Jotwani, V. et al. Tubular biomarkers and chronic kidney disease progression in SPRINT participants. Am. J. Nephrol. 51, 797–805 (2020).
Steubl, D. et al. Urinary uromodulin independently predicts end-stage renal disease and rapid kidney function decline in a cohort of chronic kidney disease patients. Medicine 98, e15808 (2019).
Kemmner, S. et al. Pretransplant serum uromodulin and its association with delayed graft function following kidney transplantation-a prospective cohort study. J. Clin. Med. 10, 2586 (2021).
Steubl, D. et al. Serum uromodulin predicts graft failure in renal transplant recipients. Biomarkers 22, 171–177 (2017).
Yazdani, B. et al. Combined use of serum uromodulin and eGFR to estimate mortality risk. Front. Med. 8, 723546 (2021).
Steubl, D. et al. Association of serum and urinary uromodulin and their correlates in older adults — The Cardiovascular Health Study. Nephrology 25, 522–526 (2020).
Delgado, G. E. et al. Serum uromodulin and mortality risk in patients undergoing coronary angiography. J. Am. Soc. Nephrol. 28, 2201–2210 (2017).
Leiherer, A. et al. Serum uromodulin is a predictive biomarker for cardiovascular events and overall mortality in coronary patients. Int. J. Cardiol. 231, 6–12 (2017).
Garimella, P. S. et al. Markers of kidney tubule function and risk of cardiovascular disease events and mortality in the SPRINT trial. Eur. Heart J. 40, 3486–3493 (2019).
Steubl, D. et al. Association of serum uromodulin with mortality and cardiovascular disease in the elderly — the Cardiovascular Health Study. Nephrol. Dial. Transpl. 35, 1399–1405 (2020).
Safirstein, R. Gene expression in nephrotoxic and ischemic acute renal failure. J. Am. Soc. Nephrol. 4, 1387–1395 (1994).
Safirstein, R. L. Acute renal failure: from renal physiology to the renal transcriptome. Kidney Int. Suppl. S62–S66 https://doi.org/10.1111/j.1523-1755.2004.09110.x (2004).
El-Achkar, T. M. et al. Tamm-Horsfall protein protects the kidney from ischemic injury by decreasing inflammation and altering TLR4 expression. Am. J. Physiol. Renal Physiol. 295, F534–F544 (2008).
Köttgen, A. et al. Multiple loci associated with indices of renal function and chronic kidney disease. Nat. Genet. 41, 712–717 (2009). GWAS that identified the link between UMOD SNPs and high CKD risk.
Pattaro, C. et al. Genome-wide association and functional follow-up reveals new loci for kidney function. PLoS Genet. 8, e1002584 (2012).
Köttgen, A. et al. New loci associated with kidney function and chronic kidney disease. Nat. Genet. 42, 376–384 (2010).
Chambers, J. C. et al. Genetic loci influencing kidney function and chronic kidney disease. Nat. Genet. 42, 373–375 (2010).
Pattaro, C. et al. A meta-analysis of genome-wide data from five European isolates reveals an association of COL22A1, SYT1, and GABRR2 with serum creatinine level. BMC Med. Genet. 11, 41 (2010).
Liu, C.-T. et al. Genetic association for renal traits among participants of African ancestry reveals new loci for renal function. PLoS Genet. 7, e1002264 (2011).
Okada, Y. et al. Meta-analysis identifies multiple loci associated with kidney function-related traits in east Asian populations. Nat. Genet. 44, 904–909 (2012).
Pattaro, C. et al. Genetic associations at 53 loci highlight cell types and biological pathways relevant for kidney function. Nat. Commun. 7, 10023 (2016).
Gorski, M. et al. Meta-analysis uncovers genome-wide significant variants for rapid kidney function decline. Kidney Int. 99, 926–939 (2021).
Gorski, M. et al. Genetic loci and prioritization of genes for kidney function decline derived from a meta-analysis of 62 longitudinal genome-wide association studies. Kidney Int. 102, 624–639 (2022).
Robinson-Cohen, C. et al. Genome-wide association study of CKD progression. J. Am. Soc. Nephrol. 34, 1547–1559 (2023).
Ponte, B. et al. Mendelian randomization to assess causality between uromodulin, blood pressure and chronic kidney disease. Kidney Int. 100, 1282–1291 (2021).
Ledo, N. et al. Functional genomic annotation of genetic risk loci highlights inflammation and epithelial biology networks in CKD. J. Am. Soc. Nephrol. 26, 692–714 (2015).
Wang, J. et al. UMOD polymorphisms associated with kidney function, serum uromodulin and risk of mortality among patients with chronic kidney disease, results from the C-STRIDE study. Genes 12, 1687 (2021).
Devuyst, O. et al. Autosomal dominant tubulointerstitial kidney disease. Nat. Rev. Dis. Prim. 5, 60 (2019).
Gast, C. et al. Autosomal dominant tubulointerstitial kidney disease-UMOD is the most frequent non polycystic genetic kidney disease. BMC Nephrol. 19, 301 (2018).
Groopman, E. E. et al. Diagnostic utility of exome sequencing for kidney disease. N. Engl. J. Med. 380, 142–151 (2019).
Eckardt, K.-U. et al. Autosomal dominant tubulointerstitial kidney disease: diagnosis, classification, and management-A KDIGO consensus report. Kidney Int. 88, 676–683 (2015).
Olinger, E. et al. Clinical and genetic spectra of autosomal dominant tubulointerstitial kidney disease due to mutations in UMOD and MUC1. Kidney Int. 98, 717–731 (2020).
Schaeffer, C., Merella, S., Pasqualetto, E., Lazarevic, D. & Rampoldi, L. Mutant uromodulin expression leads to altered homeostasis of the endoplasmic reticulum and activates the unfolded protein response. PLoS One 12, e0175970 (2017).
Jennings, P. et al. Membrane targeting and secretion of mutant uromodulin in familial juvenile hyperuricemic nephropathy. J. Am. Soc. Nephrol. 18, 264–273 (2007).
Bernascone, I. et al. Defective intracellular trafficking of uromodulin mutant isoforms. Traffic 7, 1567–1579 (2006).
Vylet’al, P. et al. Alterations of uromodulin biology: a common denominator of the genetically heterogeneous FJHN/MCKD syndrome. Kidney Int. 70, 1155–1169 (2006).
Kim, Y. et al. MANF stimulates autophagy and restores mitochondrial homeostasis to treat autosomal dominant tubulointerstitial kidney disease in mice. Nat. Commun. 14, 6493 (2023).
Trudu, M. et al. Early involvement of cellular stress and inflammatory signals in the pathogenesis of tubulointerstitial kidney disease due to UMOD mutations. Sci. Rep. 7, 7383 (2017).
Kemter, E. et al. Type of uromodulin mutation and allelic status influence onset and severity of uromodulin-associated kidney disease in mice. Hum. Mol. Genet. 22, 4148–4163 (2013).
Johnson, B. G. et al. Uromodulin p.Cys147Trp mutation drives kidney disease by activating ER stress and apoptosis. J. Clin. Invest. 127, 3954–3969 (2017).
Kemter, E., Fröhlich, T., Arnold, G. J., Wolf, E. & Wanke, R. Mitochondrial dysregulation secondary to endoplasmic reticulum stress in autosomal dominant tubulointerstitial kidney disease - UMOD (ADTKD-UMOD). Sci. Rep. 7, 42970 (2017).
Piret, S. E. et al. A mouse model for inherited renal fibrosis associated with endoplasmic reticulum stress. Dis. Model. Mech. 10, 773–786 (2017).
Ma, L. et al. Point mutation in D8C domain of Tamm-Horsfall protein/uromodulin in transgenic mice causes progressive renal damage and hyperuricemia. PLoS One 12, e0186769 (2017).
Bernascone, I. et al. A transgenic mouse model for uromodulin-associated kidney diseases shows specific tubulo-interstitial damage, urinary concentrating defect and renal failure. Hum. Mol. Genet. 19, 2998–3010 (2010).
Marshall, J. L. et al. High-resolution Slide-seqV2 spatial transcriptomics enables discovery of disease-specific cell neighborhoods and pathways. iScience 25, 104097 (2022).
Schaeffer, C. et al. Urinary secretion and extracellular aggregation of mutant uromodulin isoforms. Kidney Int. 81, 769–778 (2012).
Satanovskij, R. et al. A new missense mutation in UMOD gene leads to severely reduced serum uromodulin concentrations — a tool for the diagnosis of uromodulin-associated kidney disease. Clin. Biochem. 50, 155–158 (2017).
Chun, J. et al. Autosomal dominant tubulointerstitial kidney disease-uromodulin misclassified as focal segmental glomerulosclerosis or hereditary glomerular disease. Kidney Int. Rep. 5, 519–529 (2020).
Olinger, E. et al. An intermediate-effect size variant in UMOD confers risk for chronic kidney disease. Proc. Natl Acad. Sci. USA 119, e2114734119 (2022). Study raising the possibility that CKD and ADTKD might be part of a broad disease spectrum.
Srichai, M. B. et al. Apoptosis of the thick ascending limb results in acute kidney injury. J. Am. Soc. Nephrol. 19, 1538–1546 (2008).
Lake, B. B. et al. An atlas of healthy and injured cell states and niches in the human kidney. Nature 619, 585–594 (2023).
Mary, S. et al. Role of uromodulin in salt-sensitive hypertension. Hypertension 79, 2419–2429 (2022).
Padmanabhan, S. et al. Genome-wide association study of blood pressure extremes identifies variant near UMOD associated with hypertension. PLoS Genet. 6, e1001177 (2010).
Then, C. et al. Serum uromodulin is inversely associated with arterial hypertension and the vasoconstrictive prohormone CT-proET-1 in the population-based KORA F4 study. PLoS One 15, e0237364 (2020).
Kemter, E. et al. No amelioration of uromodulin maturation and trafficking defect by sodium 4-phenylbutyrate in vivo: studies in mouse models of uromodulin-associated kidney disease. J. Biol. Chem. 289, 10715–10726 (2014).
Dvela-Levitt, M. et al. Small molecule targets TMED9 and promotes lysosomal degradation to reverse proteinopathy. Cell 178, 521–535.e23 (2019).
McCallum, L. et al. Rationale and design of the genotype-blinded trial of torasemide for the treatment of hypertension (BHF UMOD). Am. J. Hypertens. 34, 92–99 (2021).
Acknowledgements
We thank all the members of the Ashkar laboratory, current and past collaborators, and other researchers in the related field. We disclose support for the research of this work from National Institute of Health, National Institute for Diabetes and Digestive and Kidney Diseases (R01DK111651 for TME and R00DK127216 for KAL), a VA Merit Award (5I01BX003935) for TME, and Takeda Science Foundation for AN.
Author information
Authors and Affiliations
Contributions
T.M.E. and A.N. researched data for the article. T.M.E., A.N. and K.A.L. made substantial contributions to discussions of the content. All authors wrote, reviewed or edited the manuscript before submission.
Corresponding author
Ethics declarations
Competing interests
The authors have two patents related to uromodulin: Modified Tamm–Horsfall protein and related compositions and methods of use (US11053290B2 for T.M.E.) and Materials and methods for quantifying precursor Tamm–Horsfall protein (WO2022155432A1 for T.M.E. and K.A.L.). The other authors declare no competing interests.
Peer review
Peer review information
Nature Reviews Nephrology thanks J. Scherberich, D. Steubl and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.
Additional information
Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Rights and permissions
About this article
Cite this article
Nanamatsu, A., de Araújo, L., LaFavers, K.A. et al. Advances in uromodulin biology and potential clinical applications. Nat Rev Nephrol (2024). https://doi.org/10.1038/s41581-024-00881-7
Accepted:
Published:
DOI: https://doi.org/10.1038/s41581-024-00881-7