Common noncoding UMOD gene variants induce salt-sensitive hypertension and kidney damage by increasing uromodulin expression

Matteo Trudu, Sylvie Janas, Chiara Lanzani, Huguette Debaix, Céline Schaeffer, Masami Ikehata, Lorena Citterio, Sylvie Demaretz, Francesco Trevisani, Giuseppe Ristagno, Bob Glaudemans, Kamel Laghmani, Giacomo Dell'Antonio, SKIPOGH team, Johannes Loffing, Maria P Rastaldi, Paolo Manunta, Olivier Devuyst, Luca Rampoldi, Murielle Bochud, Michel Burnier, Olivier Devuyst, Pierre-Yves Martin, Markus Mohaupt, Fred Paccaud, Antoinette Pechère-Bertschi, Bruno Vogt, Daniel Ackermann, Georg Ehret, Idris Guessous, Belen Ponte, Menno Pruijm, Matteo Trudu, Sylvie Janas, Chiara Lanzani, Huguette Debaix, Céline Schaeffer, Masami Ikehata, Lorena Citterio, Sylvie Demaretz, Francesco Trevisani, Giuseppe Ristagno, Bob Glaudemans, Kamel Laghmani, Giacomo Dell'Antonio, SKIPOGH team, Johannes Loffing, Maria P Rastaldi, Paolo Manunta, Olivier Devuyst, Luca Rampoldi, Murielle Bochud, Michel Burnier, Olivier Devuyst, Pierre-Yves Martin, Markus Mohaupt, Fred Paccaud, Antoinette Pechère-Bertschi, Bruno Vogt, Daniel Ackermann, Georg Ehret, Idris Guessous, Belen Ponte, Menno Pruijm

Abstract

Hypertension and chronic kidney disease (CKD) are complex traits representing major global health problems. Multiple genome-wide association studies have identified common variants in the promoter of the UMOD gene, which encodes uromodulin, the major protein secreted in normal urine, that cause independent susceptibility to CKD and hypertension. Despite compelling genetic evidence for the association between UMOD risk variants and disease susceptibility in the general population, the underlying biological mechanism is not understood. Here, we demonstrate that UMOD risk variants increased UMOD expression in vitro and in vivo. Uromodulin overexpression in transgenic mice led to salt-sensitive hypertension and to the presence of age-dependent renal lesions similar to those observed in elderly individuals homozygous for UMOD promoter risk variants. The link between uromodulin and hypertension is due to activation of the renal sodium cotransporter NKCC2. We demonstrated the relevance of this mechanism in humans by showing that pharmacological inhibition of NKCC2 was more effective in lowering blood pressure in hypertensive patients who are homozygous for UMOD promoter risk variants than in other hypertensive patients. Our findings link genetic susceptibility to hypertension and CKD to the level of uromodulin expression and uromodulin's effect on salt reabsorption in the kidney. These findings point to uromodulin as a therapeutic target for lowering blood pressure and preserving renal function.

Figures

Figure 1. Biological effect of UMOD SNP…
Figure 1. Biological effect of UMOD SNP variants
(a) Schematic representation of human UMOD gene structure. The position of top SNPs in the UMOD promoter associated with hypertension and CKD is shown. All variants are within the same linkage disequilibrium block, as shown by the LD plot (r2 values, data from HapMap CEU, release #28). Genotyped SNPs are shown in red. (b) UMOD expression levels (Real-Time qRT-PCR) on human nephrectomy samples homozygous for either the protective (n = 17) or risk (n = 27) UMOD variants. We normalized UMOD expression to NKCC2 to account for TAL content in the different renal samples. Distribution and mean values of expression levels are shown. ***P < 0.001 (Mann-Whitney test). (c) Urinary uromodulin concentrations in individuals of the SKIPOGH cohort by genotype at rs4293393 (n = 18 CC, 214 CT, 532 TT). Bars indicate average ± s.e.m. The P value reflects significant association of rs4293393 genotype with square-root-transformed daytime urinary uromodulin-to-creatinine ratio in a mixed linear model. (d) Partial alignment of human (Hs), gorilla (Gg), gibbon (Nl), macaque (Mm), cow (Bt) and dog (Cf) UMOD promoter sequences. The intensity of the blue color shading corresponds to nucleotide conservation. The rs4293393 SNP is predicted to be part of a glucocorticoid response element that is lost in the presence of the protective (C) allele. (e) Quantitative analysis of the relative effect of protective (C) and risk (T) alleles of SNP rs4293393 on the transcriptional activity of UMOD promoter in mTAL, highly differentiated mouse primary TAL cells retaining uromodulin expression, immortalized MKTAL (mouse kidney TAL) and HEK293 (human embryonic kidney) cells. A UMOD promoter fragment (3.7 kb) was cloned upstream of LUC (firefly luciferase) gene. We assessed the effect of variants at SNP rs4293393 on the risk haplotype by measuring luciferase activity. Graphs represent 4 independent experiments. Bars indicate average ± s.e.m. **P < 0.01; ***P < 0.001 (ANOVA followed by Bonferroni’s test).
Figure 2. Uromodulin overexpression leads to hypertension…
Figure 2. Uromodulin overexpression leads to hypertension and renal damage
(a) Box and whiskers plot showing the systolic BP at baseline in control and Umod transgenic mice at different age (n = 6–10 per group). Bars represent min and max values. (b) Average value and distribution of heart weight in 16 month-old control and TgUmodwt/wt mice (left panel) and heart histology showing left ventricular hypertrophy in TgUmodwt/wt mice (right panel) (Hematoxylin-Eosin; 1×). (c) Average systolic BP in 14 month-old control and TgUmodwt/wt mice on standard (1% NaCl) or low-sodium (0.01% NaCl) diet (n = 7 per group). Error bars indicate ± s.e.m. (d) Representative renal histological images of 16 month-old control and TgUmodwt/wt mice (n = 5). Control tissue displays normal features. Kidneys from TgUmodwt/wt mice show numerous dilated tubules (upper panel), mostly filled by casts (upper and lower panel). (e) Representative renal tissue from elderly subjects homozygous for the protective variants of rs4293393 and rs12917707 (left panels) showing normal interstitial compartment (upper panel) and mild focal tubular damage with increased thickness of the tubular basement membrane (lower panel) (n = 9). Renal tissue from subjects homozygous for the risk variants (right panels) displays dilated tubules, with detachment of the tubular epithelium (upper panel) and presence of tubular casts (lower panel) (n = 15) (PAS, bar = 100 μm). The graphs below d and e panels show average quantification of the histological analysis (only classes reaching statistical significance are shown). Error bars indicate ± s.d. (f) Tubular casts in renal tissue from a TgUmodwt/wt mouse are present in TALs (uromodulin-positive) or more distal tubules (uromodulin-negative) and are formed mostly by uromodulin (arrowhead) or by PAS-positive uromodulin-negative material (black arrows). Bar = 100 μm. (g) Transcript level (Real-Time qRT-PCR) of renal damage markers and chemokines in kidneys from 16 month-old mice: Lcn2 (Lipocalin 2); Kim-1 (Kidney injury molecule-1); Ccl2 (C-C motif chemokine 2); Ccl5 (C-C motif chemokine 5) (n = 5). Bars indicate average ± s.e.m. (h) Representative immunoblot showing Lcn2 and Kim-1 protein levels in kidney lysates of 16 month-old control and TgUmodwt/wt mice (n = 13 per group). Beta actin is shown as a loading control. Bars indicate average ± s.d. *P < 0.05; **P < 0.01; ***P < 0.001 determined by unpaired t test (a (left), b, c, h), ANOVA followed by Bonferroni’s test (a (right), g) or Mann-Whitney test (d, e).
Figure 3. Increased activation of Nkcc2 co-transporter…
Figure 3. Increased activation of Nkcc2 co-transporter and Spak kinase in TgUmodwt/wt mice
(a) Representative immunoblot analysis showing levels of phosphorylated (Thr96 and Thr101) and total Nkcc2 in kidney lysates from 16 month-old mice. Densitometric analysis (average ± s.d. of 4 independent experiments) shows increased p-Nkcc2 in TgUmodwt/wt mice (n = 10–13 per group). # unspecific signal. The increase of p-Nkcc2 is linearly correlated with Umod gene dosage (P < 0.01, ANOVA post test for linear trend). Calnexin was used as a loading control. (b) Transcript level of Nkcc2 (Real-Time qRT-PCR) on total kidney extracts from 16 month-old mice (n = 5). Bars indicate average ± s.e.m. (c) Change in sodium excretion (left panel) and systolic BP (right panel) in 16 month-old TgUmodwt/wt and control mice, 2 hours after treatment with furosemide (n = 8 Ctr, 6 TgUmodwt/wt (sodium excretion), 5 per group (systolic BP)). Bars indicate average ± s.e.m. (d) Representative immunoblot analysis showing levels of phosphorylated and total Nkcc2 and uromodulin in HEK293 cells stably expressing Nkcc2 and transfected with either wild type (Umod_wt) or soluble (Umod_sol) uromodulin. Beta actin was used as a loading control. Densitometric analysis (average ± s.d. of 3 independent experiments) is shown. (e) Quantification of Nkcc2 activity as assessed by dpHi dt−1. Bars indicate average ± s.e.m. (f) Representative immunoblot analysis showing levels of phosphorylated and total Spak and Osr1 in kidney lysates from 16 month-old control and TgUmodwt/wt mice (n = 4 per group). Bars indicate average ± s.d. *P < 0.05; **P < 0.01 determined by ANOVA followed by Bonferroni’s test (a, b, d, e) or unpaired t test (c, f).
Figure 4. Increased blood pressure and response…
Figure 4. Increased blood pressure and response to furosemide in hypertensive patients homozygous for UMOD risk variants
(a) Twenty-four hour ambulatory BP monitoring, average values for systolic (SBP) and diastolic (DBP) blood pressure are shown. DBP is significantly higher in hypertensive subjects homozygous for the UMOD risk genotype. (b, c) Response to furosemide treatment in hypertensive patients with different rs4293393 genotype. Changes in sodium excretion (b), and systolic (SBP) and diastolic (DBP) blood pressure (c) 4 hours after treatment are shown. The loop diuretic is more effective in lowering BP in hypertensive patients homozygous for rs4293393 risk variant, with a significant difference in the decrease of DBP and a similar trend for SBP (P = 0.06). This effect is associated with higher natriuretic effect. Bars indicate average ± s.e.m. (a, c) or ± s.d. (b) *P < 0.05 (ANOVA test).

References

    1. Kearney PM, et al. Global burden of hypertension: analysis of worldwide data. Lancet. 2005;365:217–223.
    1. Levey AS, et al. Chronic kidney disease as a global public health problem: approaches and initiatives - a position statement from Kidney Disease Improving Global Outcomes. Kidney Int. 2007;72:247–259.
    1. Padmanabhan S, et al. Genome-wide association study of blood pressure extremes identifies variant near UMOD associated with hypertension. PLoS Genet. 2010 doi:10.1371/journal.pgen.1001177.
    1. Kottgen A, et al. Multiple loci associated with indices of renal function and chronic kidney disease. Nat. Genet. 2009;41:712–717.
    1. Kottgen A, et al. New loci associated with kidney function and chronic kidney disease. Nat. Genet. 2010;42:376–384.
    1. 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. 2010 doi:10.1186/1471-2350-11-41.
    1. Gudbjartsson DF, et al. Association of variants at UMOD with chronic kidney disease and kidney stones-role of age and comorbid diseases. PLoS Genet. 2010 doi:10.1371/journal.pgen.1001039.
    1. Boger CA, et al. Association of eGFR-Related Loci Identified by GWAS with Incident CKD and ESRD. PLoS Genet. 2011 doi:10.1371/journal.pgen.1002292.
    1. Pattaro C, et al. Genome-wide association and functional follow-up reveals new loci for kidney function. PLoS Genet. 2012 doi:10.1371/journal.pgen.1002584.
    1. Lifton RP, Gharavi AG, Geller DS. Molecular mechanisms of human hypertension. Cell. 2001;104:545–556.
    1. Boyden LM, et al. Mutations in kelch-like 3 and cullin 3 cause hypertension and electrolyte abnormalities. Nature. 2012;482:98–102.
    1. Eckardt KU, et al. Evolving importance of kidney disease: from subspecialty to global health burden. Lancet. 2013 doi:10.1016/S0140-6736(13)60439-0.
    1. O’Seaghdha CM, Fox CS. Genome-wide association studies of chronic kidney disease: what have we learned? Nat. Rev. Nephrol. 2012;8:89–99.
    1. Padmanabhan S, Newton-Cheh C, Dominiczak AF. Genetic basis of blood pressure and hypertension. Trends Genet. 2012;28:397–408.
    1. Rampoldi L, Scolari F, Amoroso A, Ghiggeri G, Devuyst O. The rediscovery of uromodulin (Tamm-Horsfall protein): from tubulointerstitial nephropathy to chronic kidney disease. Kidney Int. 2011;80:338–347.
    1. Bates JM, et al. Tamm-Horsfall protein knockout mice are more prone to urinary tract infection: rapid communication. Kidney Int. 2004;65:791–797.
    1. Liu Y, et al. Progressive renal papillary calcification and ureteral stone formation in mice deficient for Tamm-Horsfall protein. Am. J. Physiol. Renal Physiol. 2010;299:F469–478.
    1. Bachmann S, et al. Renal effects of Tamm-Horsfall protein (uromodulin) deficiency in mice. Am. J. Physiol. Renal Physiol. 2005;288:F559–567.
    1. 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. 2011;286:30200–30210.
    1. Renigunta A, et al. Tamm-Horsfall glycoprotein interacts with renal outer medullary potassium channel ROMK2 and regulates its function. J. Biol. Chem. 2011;286:2224–2235.
    1. Hart TC, 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. Eddy AA. Scraping fibrosis: UMODulating renal fibrosis. Nat. Med. 2011;17:553–555.
    1. 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. 2010;19:2998–3010.
    1. Simpson RU, Hershey SH, Nibbelink KA. Characterization of heart size and blood pressure in the vitamin D receptor knockout mouse. J. Steroid Biochem. Mol. Bio. 2007;103:521–524.
    1. Han J, et al. Age-related changes in blood pressure in the senescence-accelerated mouse (SAM): aged SAMP1 mice manifest hypertensive vascular disease. Lab. Anim. Sci. 1998;48:256–263.
    1. Schreiber A, et al. Transcutaneous measurement of renal function in conscious mice. Am. J. Physiol. Renal Physiol. 2012;303:F783–F788.
    1. Giménez I, Forbush B. Regulatory phosphorylation sites in the NH2 terminus of the renal Na-K-Cl cotransporter (NKCC2) Am. J. Physiol. Renal Physiol. 2005;289:F1341–F1345.
    1. Ponce-Coria J, et al. Regulation of NKCC2 by a chloride-sensing mechanism involving the WNK3 and SPAK kinases. Proc. Natl. Acad. Sci. U.S.A. 2008;105:8458–8463.
    1. 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. 2009;20:589–599.
    1. Piechotta K, Lu J, Delpire E. Cation chloride cotransporters interact with the stress-related kinases Ste20-related proline-alanine-rich kinase (SPAK) and oxidative stress response 1 (OSR1) J. Biol. Chem. 2002;277:50812–50819.
    1. Richardson C, et al. Regulation of the NKCC2 ion cotransporter by SPAK-OSR1-dependent and -independent pathways. J. Cell. Sci. 2011;124:789–800.
    1. McCormick JA, et al. A SPAK isoform switch modulates renal salt transport and blood pressure. Cell Metab. 2011;14:352–364.
    1. Devuyst O. Salt wasting and blood pressure. Nat. Genet. 2008;40:495–496.
    1. Ying WZ, Sanders PW. Dietary salt regulates expression of Tamm-Horsfall glycoprotein in rats. Kidney Int. 1998;54:1150–1156.
    1. Torffvit O, Melander O, Hulten UL. Urinary excretion rate of Tamm-Horsfall protein is related to salt intake in humans. Nephron Physiol. 2004;97:31–36.
    1. Stubbe J, Madsen K, Nielsen FT, Skott O, Jensen BL. Glucocorticoid impairs growth of kidney outer medulla and accelerates loop of Henle differentiation and urinary concentrating capacity in rat kidney development. Am. J. Physiol. Renal Physiol. 2006;291:F812–822.
    1. Ferrandi M, et al. alpha- and beta-Adducin polymorphisms affect podocyte proteins and proteinuria in rodents and decline of renal function in human IgA nephropathy. J. Mol. Med. 2010;88:203–217.
    1. Mulder WJ, Hillen HF. Renal function and renal disease in the elderly: Part I. Eur. J. Intern. Med. 2001;12:86–97.
    1. Chen G, et al. Increased susceptibility of aging kidney to ischemic injury: identification of candidate genes changed during aging, but corrected by caloric restriction. Am. J. Physiol. Renal Physiol. 2007;293:F1272–1281.
    1. Genovese G, et al. Association of trypanolytic ApoL1 variants with kidney disease in African Americans. Science. 2010;329:841–845.
    1. Manunta P, et al. Physiological interaction between alpha-adducin and WNK1-NEDD4L pathways on sodium-related blood pressure regulation. Hypertension. 2008;52:366–372.
    1. Pruijm M, et al. Heritability, determinants and reference values of renal length: a family-based population study. Eur. Radiol. doi:10.1007/s00330-013-2900-4.
    1. K/DOQI clinical practice guidelines for chronic kidney disease: evaluation, classification, and stratification. Am. J. Kidney Dis. 2002;39:S1–266.
    1. Youhanna S, et al. Determination of uromodulin in human urine: influence of storage and processing. Nephrol. Dial. Transplant. 2013 Accepted for publication in.
    1. Dahan K, 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. Barr DB, et al. Urinary creatinine concentrations in the U.S. population: implications for urinary biologic monitoring measurements. Environ. Health Perspect. 2005;113:192–200.
    1. Bourgeois S, et al. Differentiated thick ascending limb (TAL) cultured cells derived from SV40 transgenic mice express functional apical NHE2 isoform: effect of nitric oxide. Pflugers Arch. 2003;446:672–683.
    1. Terryn S. A primary culture of mouse proximal tubular cells, established on collagen-coated membranes. Am. J. Physiol. Renal. Physiol. 2007;293:F476–F485.
    1. Glaudemans B, et al. A primary culture system of mouse thick ascending limb cells with preserved function and uromodulin processing. Pflugers Arch. 2013 doi: 10.1007/s00424-013-1321-1.
    1. Zaarour N, Demaretz S, Defontaine N, Mordasini D, Laghmani K. A highly conserved motif at the COOH terminus dictates endoplasmic reticulum exit and cell surface expression of NKCC2. J. Biol. Chem. 2009;284:21752–21764.
    1. Rozen S, Skaletsky H. Primer3 on the WWW for general users and for biologist programmers. Methods Mol. Biol. 2000;132:365–386.
    1. Pfaffl MW. A new mathematical model for relative quantification in real-time RT-PCR. Nucleic Acids Res. 2001;29:e45.
    1. Hummon AB, Lim SR, Difilippantonio MJ, Ried T. Isolation and solubilization of proteins after TRIzol extraction of RNA and DNA from patient material following prolonged storage. Biotechniques. 2007;42:467–470. 472.
    1. Sorensen MV, et al. Rapid dephosphorylation of the renal sodium chloride cotransporter in response to oral potassium intake in mice. Kidney Int. 2013;83:811–824.
    1. Flemmer AW, Gimenez I, Dowd BFX, Darman RB, Forbush B. Activation of the Na-K-Cl cotransporter NKCC1 detected with a phospho-specific antibody. J. Biol. Chem. 2002;277:37551–37558.
    1. Rafiqi FH, et al. Role of the WNK-activated SPAK kinase in regulating blood pressure. EMBO Mol. Med. 2010;2:63–75.
    1. Wagner CA, et al. Mouse model of type II Bartter’s syndrome. II. Altered expression of renal sodium- and water-transporting proteins. Am. J. Physiol. Renal Physiol. 2008;294:F1373–F1380.
    1. Waterhouse AM, Procter JB, Martin DM, Clamp M, Barton GJ. Jalview Version 2--a multiple sequence alignment editor and analysis workbench. Bioinformatics. 2009;25:1189–1191.
    1. Kaplan MR, Plotkin MD, Brown D, Hebert SC, Delpire E. Expression of the mouse Na-K-2Cl cotransporter, mBSC2, in the terminal inner medullary collecting duct, the glomerular and extraglomerular mesangium, and the glomerular afferent arteriole. J. Clin. Invest. 1996;98:723–730.

Source: PubMed

3
Subskrybuj