Skip to main content
Top
Published in: Current Rheumatology Reports 2/2012

01-04-2012 | CRYSTAL ARTHRITIS (MH PILLINGER, SECTION EDITOR)

Regulation of Uric Acid Excretion by the Kidney

Author: Michael S. Lipkowitz

Published in: Current Rheumatology Reports | Issue 2/2012

Login to get access

Abstract

It has been known for many years that the kidney plays a major role in uric acid homeostasis, as more than 70% of urate excretion is renal. Furthermore, hyperuricemia in gout is most commonly the result of relative urate underexcretion, as the kidney has enormous capacity for urate reabsorption. A clear understanding of the mechanisms of renal handling of urate has been hampered by the differences between humans and animal models. The power of human genetics and genome-wide association studies has now provided new insight into the molecular mechanisms of urate transport by identifying the transporters that have critical roles in urate transport. This review surveys the new evidence for a molecular model of urate transport in the renal proximal tubule and uses these data to refute the popular four-component model for urate transport that has long been in vogue. It also discusses data that help us understand the relation of diuretics to hyperuricemia, losartan-induced uricosuria, variations in uric acid levels in hyperglycemia, and the effects of dairy diets on serum urate levels. In the end, several of these clinical findings are explained, and the remaining gaps in our knowledge will become evident.
Literature
1.
2.
go back to reference Mazzali M, Hughes J, Kim YG, Jefferson JA, et al. Elevated uric acid increases blood pressure in the rat by a novel crystal-independent mechanism. Hypertension. 2001;38:1101–6.PubMedCrossRef Mazzali M, Hughes J, Kim YG, Jefferson JA, et al. Elevated uric acid increases blood pressure in the rat by a novel crystal-independent mechanism. Hypertension. 2001;38:1101–6.PubMedCrossRef
3.
go back to reference Mazzali M, Kanellis J, Han L, Feng L, et al. Hyperuricemia induces a primary renal arteriolopathy in rats by a blood pressure-independent mechanism. Am J Physiol Ren Physiol. 2002;282:F991–997. Mazzali M, Kanellis J, Han L, Feng L, et al. Hyperuricemia induces a primary renal arteriolopathy in rats by a blood pressure-independent mechanism. Am J Physiol Ren Physiol. 2002;282:F991–997.
4.
go back to reference Shi Y, Evans JE, Rock KL. Molecular identification of a danger signal that alerts the immune system to dying cells. Nature. 2003;425:516–21.PubMedCrossRef Shi Y, Evans JE, Rock KL. Molecular identification of a danger signal that alerts the immune system to dying cells. Nature. 2003;425:516–21.PubMedCrossRef
5.
go back to reference Maesaka JK, Fishbane S. Regulation of renal urate excretion: a critical review. Am J Kidney Dis. 1998;32:917–33.PubMedCrossRef Maesaka JK, Fishbane S. Regulation of renal urate excretion: a critical review. Am J Kidney Dis. 1998;32:917–33.PubMedCrossRef
6.
go back to reference Levinson DJ, Sorensen LB. Renal handling of uric acid in normal and gouty subject: evidence for a 4-component system. Ann Rheum Dis. 1980;39:173–9.PubMedCrossRef Levinson DJ, Sorensen LB. Renal handling of uric acid in normal and gouty subject: evidence for a 4-component system. Ann Rheum Dis. 1980;39:173–9.PubMedCrossRef
7.
go back to reference Abramson RG, Levitt MF. Use of pyrazinamide to assess renal uric acid transport in the rat: a micropuncture study. Am J Physiol. 1976;230:1276–83.PubMed Abramson RG, Levitt MF. Use of pyrazinamide to assess renal uric acid transport in the rat: a micropuncture study. Am J Physiol. 1976;230:1276–83.PubMed
8.
go back to reference Podevin R, Ardaillou R, Paillard F, Fontanelle J, et al. Study in man of the kinetics of the appearance of uric acid 2–14 C in the urine. Nephron. 1968;5:134–40.PubMedCrossRef Podevin R, Ardaillou R, Paillard F, Fontanelle J, et al. Study in man of the kinetics of the appearance of uric acid 2–14 C in the urine. Nephron. 1968;5:134–40.PubMedCrossRef
9.
go back to reference Gutman AB, Yu T-F. Renal function in gout. With a commentary on the renal regulation of urate excretion, and the role of the kidney in the pathogenesis of gout. Am J Med. 1957;23:600–22.PubMedCrossRef Gutman AB, Yu T-F. Renal function in gout. With a commentary on the renal regulation of urate excretion, and the role of the kidney in the pathogenesis of gout. Am J Med. 1957;23:600–22.PubMedCrossRef
10.
11.
go back to reference Praetorius E, Kirk JE. Hypouricemia: with evidence for tubular elimination of uric acid. J Lab Clin Med. 1950;35:856–68. Praetorius E, Kirk JE. Hypouricemia: with evidence for tubular elimination of uric acid. J Lab Clin Med. 1950;35:856–68.
12.
go back to reference Matsuo H, Chiba T, Nagamori S, Nakayama A, et al. Mutations in glucose transporter 9 gene SLC2A9 cause renal hypouricemia. Am J Hum Genet. 2008;83:744–51.PubMedCrossRef Matsuo H, Chiba T, Nagamori S, Nakayama A, et al. Mutations in glucose transporter 9 gene SLC2A9 cause renal hypouricemia. Am J Hum Genet. 2008;83:744–51.PubMedCrossRef
13.
go back to reference Dinour D, Gray NK, Ganon L, Knox AJ, et al. Two novel homozygous SLC2A9 mutations cause renal hypouricemia type 2. Nephrol Dial Transplant. 2011. Dinour D, Gray NK, Ganon L, Knox AJ, et al. Two novel homozygous SLC2A9 mutations cause renal hypouricemia type 2. Nephrol Dial Transplant. 2011.
14.
go back to reference Dinour D, Gray NK, Campbell S, Shu X, et al. Homozygous SLC2A9 mutations cause severe renal hypouricemia. J Am Soc Nephrol. 2010;21:64–72.PubMedCrossRef Dinour D, Gray NK, Campbell S, Shu X, et al. Homozygous SLC2A9 mutations cause severe renal hypouricemia. J Am Soc Nephrol. 2010;21:64–72.PubMedCrossRef
15.
go back to reference Steele TH. Urate secretion in man: the pyrazinamide suppression test. Ann Intern Med. 1973;79:734–7.PubMed Steele TH. Urate secretion in man: the pyrazinamide suppression test. Ann Intern Med. 1973;79:734–7.PubMed
16.
go back to reference Anzai N, Ichida K, Jutabha P, Kimura T, et al. Plasma urate level is directly regulated by a voltage-driven urate efflux transporter URATv1 (SLC2A9) in humans. J Biol Chem. 2008;283:26834–8.PubMedCrossRef Anzai N, Ichida K, Jutabha P, Kimura T, et al. Plasma urate level is directly regulated by a voltage-driven urate efflux transporter URATv1 (SLC2A9) in humans. J Biol Chem. 2008;283:26834–8.PubMedCrossRef
17.
go back to reference Steele TH, Oppenheimer S. Factors affecting urate excretion following diuretic administration in man. Am J Med. 1969;47:564–74.PubMedCrossRef Steele TH, Oppenheimer S. Factors affecting urate excretion following diuretic administration in man. Am J Med. 1969;47:564–74.PubMedCrossRef
18.
go back to reference Whitehead TP, Jungner I, Robinson D, Kolar W, et al. Serum urate, serum glucose and diabetes. Ann Clin Biochem. 1992;29:159–61.PubMed Whitehead TP, Jungner I, Robinson D, Kolar W, et al. Serum urate, serum glucose and diabetes. Ann Clin Biochem. 1992;29:159–61.PubMed
19.
go back to reference Skeith MD, Healey LA, Cutler RE. Urate excretion during mannitol and glucose diuresis. J Lab Clin Med. 1967;70:213–20.PubMed Skeith MD, Healey LA, Cutler RE. Urate excretion during mannitol and glucose diuresis. J Lab Clin Med. 1967;70:213–20.PubMed
20.
go back to reference Bailey CJ, Gross JL, Pieters A, Bastien A, et al. Effect of dapagliflozin in patients with type 2 diabetes who have inadequate glycaemic control with metformin: a randomised, double-blind, placebo-controlled trial. Lancet. 375: 2223–33. Bailey CJ, Gross JL, Pieters A, Bastien A, et al. Effect of dapagliflozin in patients with type 2 diabetes who have inadequate glycaemic control with metformin: a randomised, double-blind, placebo-controlled trial. Lancet. 375: 2223–33.
21.
go back to reference Dalbeth N, Wong S, Gamble GD, Horne A, et al. Acute effect of milk on serum urate concentrations: a randomised controlled crossover trial. Ann Rheum Dis. 69:1677–82. Dalbeth N, Wong S, Gamble GD, Horne A, et al. Acute effect of milk on serum urate concentrations: a randomised controlled crossover trial. Ann Rheum Dis. 69:1677–82.
22.
go back to reference Choi HK, Atkinson K, Karlson EW, Willett W, et al. Purine-rich foods, dairy and protein intake, and the risk of gout in men. N Engl J Med. 2004;350:1093–103.PubMedCrossRef Choi HK, Atkinson K, Karlson EW, Willett W, et al. Purine-rich foods, dairy and protein intake, and the risk of gout in men. N Engl J Med. 2004;350:1093–103.PubMedCrossRef
23.
go back to reference Dolaeus J, Stephens W. Dolaeus Upon the Cure of the Gout by Milk-Diet. To which is prefixed an Essay upon diet. By William Stephens, M.D. F.R.S. Fellow of the King and Queen’s College of Physicians in Ireland, Physician to the Royal Hospital, and Botany Lecturer in the University of Dublin. Printed for J. Smith and W. Bruce on the Blind-Key in Dublin: And Sold by John Osborn and Thomas Longman in Pater-noster-Row, 1732. Dolaeus J, Stephens W. Dolaeus Upon the Cure of the Gout by Milk-Diet. To which is prefixed an Essay upon diet. By William Stephens, M.D. F.R.S. Fellow of the King and Queen’s College of Physicians in Ireland, Physician to the Royal Hospital, and Botany Lecturer in the University of Dublin. Printed for J. Smith and W. Bruce on the Blind-Key in Dublin: And Sold by John Osborn and Thomas Longman in Pater-noster-Row, 1732.
24.
go back to reference Burnier M, Roch-Ramel F, Brunner HR. Renal effects of angiotensin II receptor blockade in normotensive subjects. Kidney Int. 1996;49:1787–90.PubMedCrossRef Burnier M, Roch-Ramel F, Brunner HR. Renal effects of angiotensin II receptor blockade in normotensive subjects. Kidney Int. 1996;49:1787–90.PubMedCrossRef
25.
go back to reference Edwards RM, Trizna W, Stack EJ, Weinstock J. Interaction of nonpeptide angiotensin II receptor antagonists with the urate transporter in rat renal brush-border membranes. J Pharmacol Exp Ther. 1996;276:125–9.PubMed Edwards RM, Trizna W, Stack EJ, Weinstock J. Interaction of nonpeptide angiotensin II receptor antagonists with the urate transporter in rat renal brush-border membranes. J Pharmacol Exp Ther. 1996;276:125–9.PubMed
26.
go back to reference Rafey MA, Lipkowitz MS, Leal-Pinto E, Abramson RG. Uric acid transport. Curr Opin Nephrol Hypertens. 2003;12:511–6.PubMedCrossRef Rafey MA, Lipkowitz MS, Leal-Pinto E, Abramson RG. Uric acid transport. Curr Opin Nephrol Hypertens. 2003;12:511–6.PubMedCrossRef
27.
go back to reference Abramson RG, Lipkowitz MS. Evolution of the uric acid transport mechanisms in vertebrate kidney. In: Kinne RKH, editor. Basic principles in transport, vol. 3. Basel: Karger; 1990. p. 115–53. Abramson RG, Lipkowitz MS. Evolution of the uric acid transport mechanisms in vertebrate kidney. In: Kinne RKH, editor. Basic principles in transport, vol. 3. Basel: Karger; 1990. p. 115–53.
28.
go back to reference Wu XW, Muzny DM, Lee CC, Caskey CT. Two independent mutational events in the loss of urate oxidase during hominoid evolution. J Mol Evol. 1992;34:78–84.PubMedCrossRef Wu XW, Muzny DM, Lee CC, Caskey CT. Two independent mutational events in the loss of urate oxidase during hominoid evolution. J Mol Evol. 1992;34:78–84.PubMedCrossRef
29.
go back to reference Wu X, Wakamiya M, Vaishnav S, Geske R, et al. Hyperuricemia and urate nephropathy in urate oxidase-deficient mice. Proc Natl Acad Sci U S A. 1994;91:742–6.PubMedCrossRef Wu X, Wakamiya M, Vaishnav S, Geske R, et al. Hyperuricemia and urate nephropathy in urate oxidase-deficient mice. Proc Natl Acad Sci U S A. 1994;91:742–6.PubMedCrossRef
30.
go back to reference Enomoto A, Kimura H, Chairoungdua A, Shigeta Y, et al. Molecular identification of a renal urate anion exchanger that regulates blood urate levels. Nature. 2002;417:447–52.PubMed Enomoto A, Kimura H, Chairoungdua A, Shigeta Y, et al. Molecular identification of a renal urate anion exchanger that regulates blood urate levels. Nature. 2002;417:447–52.PubMed
31.
go back to reference Guggino SE, Aronson PS. Paradoxical effects of pyrazinoate and nicotinate on urate transport in dog renal microvillus membranes. J Clin Invest. 1985;76:543–7.PubMedCrossRef Guggino SE, Aronson PS. Paradoxical effects of pyrazinoate and nicotinate on urate transport in dog renal microvillus membranes. J Clin Invest. 1985;76:543–7.PubMedCrossRef
32.
go back to reference Guggino SE, Martin GJ, Aronson PS. Specificity and modes of the anion exchanger in dog renal microvillus membranes. Am J Physiol. 1983;244:F612–21.PubMed Guggino SE, Martin GJ, Aronson PS. Specificity and modes of the anion exchanger in dog renal microvillus membranes. Am J Physiol. 1983;244:F612–21.PubMed
33.
go back to reference Jang WC, Nam YH, Ahn YC, Park SM, et al. G109T polymorphism of SLC22A12 gene is associated with serum uric acid level, but not with metabolic syndrome. Rheumatol Int. 2011. Jang WC, Nam YH, Ahn YC, Park SM, et al. G109T polymorphism of SLC22A12 gene is associated with serum uric acid level, but not with metabolic syndrome. Rheumatol Int. 2011.
34.
go back to reference Dinour D, Bahn A, Ganon L, Ron R, et al. URAT1 mutations cause renal hypouricemia type 1 in Iraqi Jews. Nephrol Dial Transplant. 2011;26:2175–81.PubMedCrossRef Dinour D, Bahn A, Ganon L, Ron R, et al. URAT1 mutations cause renal hypouricemia type 1 in Iraqi Jews. Nephrol Dial Transplant. 2011;26:2175–81.PubMedCrossRef
35.
go back to reference Kenny EE, Kim M, Gusev A, Lowe JK, et al. Increased power of mixed models facilitates association mapping of 10 loci for metabolic traits in an isolated population. Hum Mol Genet. 2011;20:827–39.PubMedCrossRef Kenny EE, Kim M, Gusev A, Lowe JK, et al. Increased power of mixed models facilitates association mapping of 10 loci for metabolic traits in an isolated population. Hum Mol Genet. 2011;20:827–39.PubMedCrossRef
36.
go back to reference Kolz M, Johnson T, Sanna S, Teumer A, et al. Meta-analysis of 28,141 individuals identifies common variants within five new loci that influence uric acid concentrations. PLoS Genet. 2009;5:e1000504.PubMedCrossRef Kolz M, Johnson T, Sanna S, Teumer A, et al. Meta-analysis of 28,141 individuals identifies common variants within five new loci that influence uric acid concentrations. PLoS Genet. 2009;5:e1000504.PubMedCrossRef
37.
go back to reference Komoda F, Sekine T, Inatomi J, Enomoto A, et al. The W258X mutation in SLC22A12 is the predominant cause of Japanese renal hypouricemia. Pediatr Nephrol. 2004;19:728–33.PubMedCrossRef Komoda F, Sekine T, Inatomi J, Enomoto A, et al. The W258X mutation in SLC22A12 is the predominant cause of Japanese renal hypouricemia. Pediatr Nephrol. 2004;19:728–33.PubMedCrossRef
39.
go back to reference Dehghan A, Kottgen A, Yang Q, Hwang SJ, et al. Association of three genetic loci with uric acid concentration and risk of gout: a genome-wide association study. Lancet. 2008;372:1953–61.PubMedCrossRef Dehghan A, Kottgen A, Yang Q, Hwang SJ, et al. Association of three genetic loci with uric acid concentration and risk of gout: a genome-wide association study. Lancet. 2008;372:1953–61.PubMedCrossRef
40.
go back to reference Li S, Sanna S, Maschio A, Busonero F, et al. The GLUT9 gene is associated with serum uric acid levels in Sardinia and Chianti cohorts. PLoS Genet. 2007;3:e194.PubMedCrossRef Li S, Sanna S, Maschio A, Busonero F, et al. The GLUT9 gene is associated with serum uric acid levels in Sardinia and Chianti cohorts. PLoS Genet. 2007;3:e194.PubMedCrossRef
41.
go back to reference Wallace C, Newhouse SJ, Braund P, Zhang F, et al. Genome-wide association study identifies genes for biomarkers of cardiovascular disease: serum urate and dyslipidemia. Am J Hum Genet. 2008;82:139–49.PubMedCrossRef Wallace C, Newhouse SJ, Braund P, Zhang F, et al. Genome-wide association study identifies genes for biomarkers of cardiovascular disease: serum urate and dyslipidemia. Am J Hum Genet. 2008;82:139–49.PubMedCrossRef
42.
go back to reference Phay JE, Hussain HB, Moley JF. Cloning and expression analysis of a novel member of the facilitative glucose transporter family, SLC2A9 (GLUT9). Genomics. 2000;66:217–20.PubMedCrossRef Phay JE, Hussain HB, Moley JF. Cloning and expression analysis of a novel member of the facilitative glucose transporter family, SLC2A9 (GLUT9). Genomics. 2000;66:217–20.PubMedCrossRef
43.
go back to reference Keembiyehetty C, Augustin R, Carayannopoulos MO, Steer S, et al. Mouse glucose transporter 9 splice variants are expressed in adult liver and kidney and are up-regulated in diabetes. Mol Endocrinol. 2006;20:686–97.PubMedCrossRef Keembiyehetty C, Augustin R, Carayannopoulos MO, Steer S, et al. Mouse glucose transporter 9 splice variants are expressed in adult liver and kidney and are up-regulated in diabetes. Mol Endocrinol. 2006;20:686–97.PubMedCrossRef
44.
go back to reference Augustin R, Carayannopoulos MO, Dowd LO, Phay JE, et al. Identification and characterization of human glucose transporter-like protein-9 (GLUT9): alternative splicing alters trafficking. J Biol Chem. 2004;279:16229–36.PubMedCrossRef Augustin R, Carayannopoulos MO, Dowd LO, Phay JE, et al. Identification and characterization of human glucose transporter-like protein-9 (GLUT9): alternative splicing alters trafficking. J Biol Chem. 2004;279:16229–36.PubMedCrossRef
45.
go back to reference Caulfield MJ, Munroe PB, O’Neill D, Witkowska K, et al. SLC2A9 is a high-capacity urate transporter in humans. PLoS Med. 2008;5:e197.PubMedCrossRef Caulfield MJ, Munroe PB, O’Neill D, Witkowska K, et al. SLC2A9 is a high-capacity urate transporter in humans. PLoS Med. 2008;5:e197.PubMedCrossRef
46.
go back to reference Vitart V, Rudan I, Hayward C, Gray NK, et al. SLC2A9 is a newly identified urate transporter influencing serum urate concentration, urate excretion and gout. Nat Genet. 2008;40:437–42.PubMedCrossRef Vitart V, Rudan I, Hayward C, Gray NK, et al. SLC2A9 is a newly identified urate transporter influencing serum urate concentration, urate excretion and gout. Nat Genet. 2008;40:437–42.PubMedCrossRef
47.
go back to reference Preitner F, Bonny O, Laverriere A, Rotman S, et al. Glut9 is a major regulator of urate homeostasis and its genetic inactivation induces hyperuricosuria and urate nephropathy. Proc Natl Acad Sci U S A. 2009;106:15501–6.PubMedCrossRef Preitner F, Bonny O, Laverriere A, Rotman S, et al. Glut9 is a major regulator of urate homeostasis and its genetic inactivation induces hyperuricosuria and urate nephropathy. Proc Natl Acad Sci U S A. 2009;106:15501–6.PubMedCrossRef
48.
go back to reference Mobasheri A, Neama G, Bell S, Richardson S, et al. Human articular chondrocytes express three facilitative glucose transporter isoforms: GLUT1, GLUT3 and GLUT9. Cell Biol Int. 2002;26:297–300.PubMedCrossRef Mobasheri A, Neama G, Bell S, Richardson S, et al. Human articular chondrocytes express three facilitative glucose transporter isoforms: GLUT1, GLUT3 and GLUT9. Cell Biol Int. 2002;26:297–300.PubMedCrossRef
49.
go back to reference Yang Q, Kottgen A, Dehghan A, Smith AV, et al. Multiple genetic loci influence serum urate levels and their relationship with gout and cardiovascular disease risk factors. Circ Cardiovasc Genet. 2010;3:523–30.PubMedCrossRef Yang Q, Kottgen A, Dehghan A, Smith AV, et al. Multiple genetic loci influence serum urate levels and their relationship with gout and cardiovascular disease risk factors. Circ Cardiovasc Genet. 2010;3:523–30.PubMedCrossRef
50.
go back to reference Woodward OM, Kottgen A, Coresh J, Boerwinkle E, et al. Identification of a urate transporter, ABCG2, with a common functional polymorphism causing gout. Proc Natl Acad Sci U S A. 2009;106:10338–42.PubMedCrossRef Woodward OM, Kottgen A, Coresh J, Boerwinkle E, et al. Identification of a urate transporter, ABCG2, with a common functional polymorphism causing gout. Proc Natl Acad Sci U S A. 2009;106:10338–42.PubMedCrossRef
51.
go back to reference Yamane S, Reddi AH. Induction of chondrogenesis and superficial zone protein accumulation in synovial side population cells by BMP-7 and TGF-beta1. J Orthop Res. 2008;26:485–92.PubMedCrossRef Yamane S, Reddi AH. Induction of chondrogenesis and superficial zone protein accumulation in synovial side population cells by BMP-7 and TGF-beta1. J Orthop Res. 2008;26:485–92.PubMedCrossRef
52.
go back to reference Iharada M, Miyaji T, Fujimoto T, Hiasa M, et al. Type 1 sodium-dependent phosphate transporter (SLC17A1 protein) is a Cl(−)-dependent urate exporter. J Biol Chem. 285:26107–13. Iharada M, Miyaji T, Fujimoto T, Hiasa M, et al. Type 1 sodium-dependent phosphate transporter (SLC17A1 protein) is a Cl(−)-dependent urate exporter. J Biol Chem. 285:26107–13.
53.
go back to reference Jutabha P, Anzai N, Kitamura K, Taniguchi A, et al. Human sodium phosphate transporter 4 (hNPT4/SLC17A3) as a common renal secretory pathway for drugs and urate. J Biol Chem. 285:35123–32. Jutabha P, Anzai N, Kitamura K, Taniguchi A, et al. Human sodium phosphate transporter 4 (hNPT4/SLC17A3) as a common renal secretory pathway for drugs and urate. J Biol Chem. 285:35123–32.
54.
go back to reference Eraly SA, Vallon V, Rieg T, Gangoiti JA, et al. Multiple organic anion transporters contribute to net renal excretion of uric acid. Physiol Genom. 2008;33:180–92.CrossRef Eraly SA, Vallon V, Rieg T, Gangoiti JA, et al. Multiple organic anion transporters contribute to net renal excretion of uric acid. Physiol Genom. 2008;33:180–92.CrossRef
55.
go back to reference Xu G, Bhatnagar V, Wen G, Hamilton BA, et al. Analyses of coding region polymorphisms in apical and basolateral human organic anion transporter (OAT) genes [OAT1 (NKT), OAT2, OAT3, OAT4, URAT (RST)]. Kidney Int. 2005;68:1491–9.PubMedCrossRef Xu G, Bhatnagar V, Wen G, Hamilton BA, et al. Analyses of coding region polymorphisms in apical and basolateral human organic anion transporter (OAT) genes [OAT1 (NKT), OAT2, OAT3, OAT4, URAT (RST)]. Kidney Int. 2005;68:1491–9.PubMedCrossRef
56.
go back to reference Ekaratanawong S, Anzai N, Jutabha P, Miyazaki H, et al. Human organic anion transporter 4 is a renal apical organic anion/dicarboxylate exchanger in the proximal tubules. J Pharmacol Sci. 2004;94:297–304.PubMedCrossRef Ekaratanawong S, Anzai N, Jutabha P, Miyazaki H, et al. Human organic anion transporter 4 is a renal apical organic anion/dicarboxylate exchanger in the proximal tubules. J Pharmacol Sci. 2004;94:297–304.PubMedCrossRef
57.
go back to reference Hyink DP, Rappoport JZ, Wilson PD, Abramson RG. Expression of the urate transporter/channel is developmentally regulated in human kidneys. Am J Physiol Ren Physiol. 2001;281:F875–886. Hyink DP, Rappoport JZ, Wilson PD, Abramson RG. Expression of the urate transporter/channel is developmentally regulated in human kidneys. Am J Physiol Ren Physiol. 2001;281:F875–886.
58.
go back to reference Lipkowitz MS, Leal-Pinto E, Rappoport JZ, Najfeld V, et al. Functional reconstitution, membrane targeting, genomic structure, and chromosomal localization of a human urate transporter. J Clin Invest. 2001;107:1103–15.PubMedCrossRef Lipkowitz MS, Leal-Pinto E, Rappoport JZ, Najfeld V, et al. Functional reconstitution, membrane targeting, genomic structure, and chromosomal localization of a human urate transporter. J Clin Invest. 2001;107:1103–15.PubMedCrossRef
59.
go back to reference Leal-Pinto E, Cohen BE, Lipkowitz MS, Abramson RG. Functional analysis and molecular model of the human urate transporter/channel, hUAT. Am J Physiol Ren Physiol. 2002;283:F150–163. Leal-Pinto E, Cohen BE, Lipkowitz MS, Abramson RG. Functional analysis and molecular model of the human urate transporter/channel, hUAT. Am J Physiol Ren Physiol. 2002;283:F150–163.
60.
go back to reference Lipkowitz MS, Leal-Pinto E, Cohen BE, Abramson RG. Galectin 9 is the sugar-regulated urate transporter/channel UAT. Glycoconj J. 2004;19:491–8.PubMedCrossRef Lipkowitz MS, Leal-Pinto E, Cohen BE, Abramson RG. Galectin 9 is the sugar-regulated urate transporter/channel UAT. Glycoconj J. 2004;19:491–8.PubMedCrossRef
61.
go back to reference Van Aubel RA, Smeets PH, van den Heuvel JJ, Russel FG. Human organic anion transporter MRP4 (ABCC4) is an efflux pump for the purine end metabolite urate with multiple allosteric substrate binding sites. Am J Physiol Ren Physiol. 2005;288:F327–333.CrossRef Van Aubel RA, Smeets PH, van den Heuvel JJ, Russel FG. Human organic anion transporter MRP4 (ABCC4) is an efflux pump for the purine end metabolite urate with multiple allosteric substrate binding sites. Am J Physiol Ren Physiol. 2005;288:F327–333.CrossRef
62.
go back to reference Gopal E, Umapathy NS, Martin PM, Ananth S, et al. Cloning and functional characterization of human SMCT2 (SLC5A12) and expression pattern of the transporter in kidney. Biochim Biophys Acta. 2007;1768:2690–7.PubMedCrossRef Gopal E, Umapathy NS, Martin PM, Ananth S, et al. Cloning and functional characterization of human SMCT2 (SLC5A12) and expression pattern of the transporter in kidney. Biochim Biophys Acta. 2007;1768:2690–7.PubMedCrossRef
63.
go back to reference Anzai N, Miyazaki H, Noshiro R, Khamdang S, et al. The multivalent PDZ domain-containing protein PDZK1 regulates transport activity of renal urate-anion exchanger URAT1 via its C terminus. J Biol Chem. 2004;279:45942–50.PubMedCrossRef Anzai N, Miyazaki H, Noshiro R, Khamdang S, et al. The multivalent PDZ domain-containing protein PDZK1 regulates transport activity of renal urate-anion exchanger URAT1 via its C terminus. J Biol Chem. 2004;279:45942–50.PubMedCrossRef
64.
go back to reference Jutabha P, Anzai N, Endou H. Interaction of the multivalent PDZ damain protein PDZK1 with type 1 sodium-phosphate cotransporter (NPT1). J Am Soc Nephrol. 2005;16:350A. Jutabha P, Anzai N, Endou H. Interaction of the multivalent PDZ damain protein PDZK1 with type 1 sodium-phosphate cotransporter (NPT1). J Am Soc Nephrol. 2005;16:350A.
65.
go back to reference Miyazaki H, Anzai N, Ekaratanawong S, Sakata T, et al. Modulation of renal apical organic anion transporter 4 function by two PDZ domain-containing proteins. J Am Soc Nephrol. 2005;16:3498–506.PubMedCrossRef Miyazaki H, Anzai N, Ekaratanawong S, Sakata T, et al. Modulation of renal apical organic anion transporter 4 function by two PDZ domain-containing proteins. J Am Soc Nephrol. 2005;16:3498–506.PubMedCrossRef
66.
go back to reference Halestrap AP, Price NT. The proton-linked monocarboxylate transporter (MCT) family: structure, function and regulation. Biochem J. 1999;343(Pt 2):281–99.PubMedCrossRef Halestrap AP, Price NT. The proton-linked monocarboxylate transporter (MCT) family: structure, function and regulation. Biochem J. 1999;343(Pt 2):281–99.PubMedCrossRef
67.
go back to reference Jutabha P, Anzai N, Kitamura K, Taniguchi A, et al. Human sodium phosphate transporter 4 (hNPT4/SLC17A3) as a common renal secretory pathway for drugs and urate. J Biol Chem. 2011;285:35123–32.CrossRef Jutabha P, Anzai N, Kitamura K, Taniguchi A, et al. Human sodium phosphate transporter 4 (hNPT4/SLC17A3) as a common renal secretory pathway for drugs and urate. J Biol Chem. 2011;285:35123–32.CrossRef
68.
go back to reference Dalbeth N, Wong S, Gamble GD, Horne A, et al. Acute effect of milk on serum urate concentrations: a randomised controlled crossover trial. Ann Rheum Dis. 2010;69:1677–82.PubMedCrossRef Dalbeth N, Wong S, Gamble GD, Horne A, et al. Acute effect of milk on serum urate concentrations: a randomised controlled crossover trial. Ann Rheum Dis. 2010;69:1677–82.PubMedCrossRef
Metadata
Title
Regulation of Uric Acid Excretion by the Kidney
Author
Michael S. Lipkowitz
Publication date
01-04-2012
Publisher
Current Science Inc.
Published in
Current Rheumatology Reports / Issue 2/2012
Print ISSN: 1523-3774
Electronic ISSN: 1534-6307
DOI
https://doi.org/10.1007/s11926-012-0240-z

Other articles of this Issue 2/2012

Current Rheumatology Reports 2/2012 Go to the issue

CRYSTAL ARTHRITIS (MH PILLINGER, SECTION EDITOR)

Axial (Spinal) Gout

CRYSTAL ARTHRITIS (MH PILLINGER, SECTION EDITOR)

Gout and Organ Transplantation