Skip to main content
Top
Published in: BMC Nephrology 1/2017

Open Access 01-12-2017 | Research article

Uric acid causes kidney injury through inducing fibroblast expansion, Endothelin-1 expression, and inflammation

Authors: Muhammad Mansyur Romi, Nur Arfian, Untung Tranggono, Wiwit Ananda Wahyu Setyaningsih, Dwi Cahyani Ratna Sari

Published in: BMC Nephrology | Issue 1/2017

Login to get access

Abstract

Background

Uric acid (UA) plays important roles in inducing renal inflammation, intra-renal vasoconstriction and renal damage. Endothelin-1 (ET-1) is a well-known profibrotic factor in the kidney and is associated with fibroblast expansion. We examined the role of hyperuricemia conditions in causing elevation of ET-1 expression and kidney injury.

Methods

Hyperuricemia was induced in mice using daily intraperitoneal injection of uric acid 125 mg/Kg body weight. An NaCl injection was used in control mice. Mice were euthanized on days-7 (UA7) and 14 (UA14). We also added allopurinol groups (UAL7 and UAL14) with supplementation of allopurinol 50 mg/Kg body weight orally. Uric acid and creatinine serum were measured from blood serum. Periodic Acid Schiff (PAS) and Sirius Red staining were done for glomerulosclerosis, tubular injury and fibrosis quantification. mRNA expression examination was performed for nephrin, podocin, preproEndothelin-1 (ppET-1), MCP-1 and ICAM-1. PDGFRβ immunostaining was done for quantification of fibroblast, while α-SMA immunostaining was done for localizing myofibroblast. Western blot analysis was conducted to quantify TGF-β1, α-SMA and Endothelin A Receptor (ETAR) protein expression.

Results

Uric acid and creatinine levels were elevated after 7 and 14 days and followed by significant increase of glomerulosclerosis and tubular injury score in the uric acid group (p < 0.05 vs. control). Both UA7 and UA14 groups had higher fibrosis, tubular injury and glomerulosclerosis with significant increase of fibroblast cell number compared with control. RT-PCR revealed down-regulation of nephrin and podocin expression (p < 0.05 vs. control), and up-regulation of MCP-1, ET-1 and ICAM-1 expression (p < 0.05 vs. control). Western blot revealed higher expression of TGF-β1 and α-SMA protein expression. Determination of allopurinol attenuated kidney injury was based on reduction of fibroblast cell number, inflammation mediators and ppET-1 expression with reduction of TGF-β1 and α-SMA protein expression.

Conclusions

UA induced glomerulosclerosis, tubular injury and renal fibrosis with reduction of podocyte function and inflammatory mediator elevation. ET-1 and fibroblast expansion might modulate hyperuricemia induced renal fibrosis.
Literature
1.
go back to reference Neuhofer W, Pittrow D. Role of endothelin and endothelin receptor antagonists in renal disease. Eur J Clin Investig. 2006;36(Suppl 3):78–88.CrossRef Neuhofer W, Pittrow D. Role of endothelin and endothelin receptor antagonists in renal disease. Eur J Clin Investig. 2006;36(Suppl 3):78–88.CrossRef
2.
go back to reference Kang DH, Nakagawa T, et al. A role for uric acid in the progression of renal disease. J Am Soc Nephrol. 2002;13(12):2888–97.CrossRefPubMed Kang DH, Nakagawa T, et al. A role for uric acid in the progression of renal disease. J Am Soc Nephrol. 2002;13(12):2888–97.CrossRefPubMed
3.
go back to reference Kanellis J, Kang DH. Uric acid as a mediator of endothelial dysfunction, inflammation, and vascular disease. Semin Nephrol. 2005;25(1):39–42.CrossRefPubMed Kanellis J, Kang DH. Uric acid as a mediator of endothelial dysfunction, inflammation, and vascular disease. Semin Nephrol. 2005;25(1):39–42.CrossRefPubMed
4.
go back to reference Wu XW, Muzny DM, et al. Two independent mutational events in the loss of urate oxidase during hominoid evolution. J Mol Evol. 1992;34(1):78–84.CrossRefPubMed Wu XW, Muzny DM, et al. Two independent mutational events in the loss of urate oxidase during hominoid evolution. J Mol Evol. 1992;34(1):78–84.CrossRefPubMed
5.
go back to reference Johnson RJ, Kang DH, et al. Is there a pathogenetic role for uric acid in hypertension and cardiovascular and renal disease? Hypertension. 2003;41(6):1183–90.CrossRefPubMed Johnson RJ, Kang DH, et al. Is there a pathogenetic role for uric acid in hypertension and cardiovascular and renal disease? Hypertension. 2003;41(6):1183–90.CrossRefPubMed
6.
go back to reference Enomoto A, Kimura H, et al. Molecular identification of a renal urate anion exchanger that regulates blood urate levels. Nature. 2002;417(6887):447–52.CrossRefPubMed Enomoto A, Kimura H, et al. Molecular identification of a renal urate anion exchanger that regulates blood urate levels. Nature. 2002;417(6887):447–52.CrossRefPubMed
7.
go back to reference Zoccali C, Maio R, et al. Uric acid and endothelial dysfunction in essential hypertension. J Am Soc Nephrol. 2006;17(5):1466–71.CrossRefPubMed Zoccali C, Maio R, et al. Uric acid and endothelial dysfunction in essential hypertension. J Am Soc Nephrol. 2006;17(5):1466–71.CrossRefPubMed
8.
go back to reference Obermay RP, Temml C, et al. Elevated uric acid increases the risk for kidney disease. J Am Soc Nephrol. 2008;19(12):2407–13.CrossRef Obermay RP, Temml C, et al. Elevated uric acid increases the risk for kidney disease. J Am Soc Nephrol. 2008;19(12):2407–13.CrossRef
9.
go back to reference Mene P, Punzo G. Uric acid: bystander or culprit in hypertension and progressive renal disease? J Hypertens. 2008;26(11):2085–92.CrossRefPubMed Mene P, Punzo G. Uric acid: bystander or culprit in hypertension and progressive renal disease? J Hypertens. 2008;26(11):2085–92.CrossRefPubMed
10.
go back to reference Berger L, Yu TF. Renal function in gout. IV. An analysis of 524 gouty subjects including long-term follow-up studies. Am J Med. 1975;59(5):605–13.CrossRefPubMed Berger L, Yu TF. Renal function in gout. IV. An analysis of 524 gouty subjects including long-term follow-up studies. Am J Med. 1975;59(5):605–13.CrossRefPubMed
12.
go back to reference Brand FN, McGee DL, et al. Hyperuricemia as a risk factor of coronary heart disease: the Framingham study. Am J Epidemiol. 1985;121(1):11–8.CrossRefPubMed Brand FN, McGee DL, et al. Hyperuricemia as a risk factor of coronary heart disease: the Framingham study. Am J Epidemiol. 1985;121(1):11–8.CrossRefPubMed
13.
go back to reference Cannon PJ, Stason WB, et al. Hyperuricemia in primary and renal hypertension. N Engl J Med. 1966;275(9):457–64.CrossRefPubMed Cannon PJ, Stason WB, et al. Hyperuricemia in primary and renal hypertension. N Engl J Med. 1966;275(9):457–64.CrossRefPubMed
14.
go back to reference di Giovine FS, Malawista SE, et al. Urate crystals stimulate production of tumor necrosis factor alpha from human blood monocytes and synovial cells. Cytokine mRNA and protein kinetics, and cellular distribution. J Clin Invest. 1991;87(4):1375–81.CrossRefPubMedPubMedCentral di Giovine FS, Malawista SE, et al. Urate crystals stimulate production of tumor necrosis factor alpha from human blood monocytes and synovial cells. Cytokine mRNA and protein kinetics, and cellular distribution. J Clin Invest. 1991;87(4):1375–81.CrossRefPubMedPubMedCentral
15.
go back to reference Mazzali M, Kanellis J, et al. Hyperuricemia induces a primary renal arteriolopathy in rats by a blood pressure-independent mechanism. Am J Physiol Renal Physiol. 2002;282(6):F991–7.CrossRefPubMed Mazzali M, Kanellis J, et al. Hyperuricemia induces a primary renal arteriolopathy in rats by a blood pressure-independent mechanism. Am J Physiol Renal Physiol. 2002;282(6):F991–7.CrossRefPubMed
16.
go back to reference Sanchez-Lozada LG, Tapia E, et al. Mild hyperuricemia induces vasoconstriction and maintains glomerular hypertension in normal and remnant kidney rats. Kidney Int. 2005;67(1):237–47.CrossRefPubMed Sanchez-Lozada LG, Tapia E, et al. Mild hyperuricemia induces vasoconstriction and maintains glomerular hypertension in normal and remnant kidney rats. Kidney Int. 2005;67(1):237–47.CrossRefPubMed
17.
go back to reference Rao GN, Corson MA, et al. Uric acid stimulates vascular smooth muscle cell proliferation by increasing platelet-derived growth factor A-chain expression. J Biol Chem. 1991;266(13):8604–8.PubMed Rao GN, Corson MA, et al. Uric acid stimulates vascular smooth muscle cell proliferation by increasing platelet-derived growth factor A-chain expression. J Biol Chem. 1991;266(13):8604–8.PubMed
18.
go back to reference Kanellis J, Watanabe S, et al. Uric acid stimulates monocyte chemoattractant protein-1 production in vascular smooth muscle cells via mitogen-activated protein kinase and cyclooxygenase-2. Hypertension. 2003;41(6):1287–93.CrossRefPubMed Kanellis J, Watanabe S, et al. Uric acid stimulates monocyte chemoattractant protein-1 production in vascular smooth muscle cells via mitogen-activated protein kinase and cyclooxygenase-2. Hypertension. 2003;41(6):1287–93.CrossRefPubMed
19.
go back to reference Yanagisawa M, Kurihara H, et al. A novel potent vasoconstrictor peptide produced by vascular endothelial cells. Nature. 1998;332(6163):411–5.CrossRef Yanagisawa M, Kurihara H, et al. A novel potent vasoconstrictor peptide produced by vascular endothelial cells. Nature. 1998;332(6163):411–5.CrossRef
20.
go back to reference Inazaki K, Kanamaru Y, et al. Smad3 deficiency attenuates renal fibrosis, inflammation,and apoptosis after unilateral ureteral obstruction. Kidney Int. 2004;66(2):597–604.CrossRefPubMed Inazaki K, Kanamaru Y, et al. Smad3 deficiency attenuates renal fibrosis, inflammation,and apoptosis after unilateral ureteral obstruction. Kidney Int. 2004;66(2):597–604.CrossRefPubMed
21.
go back to reference Yang Z, Xiaohua W, et al. Uric acid increases fibronectin synthesis through upregulation of lysyl oxidase expression in rat renal tubular epithelial cells. Am J Physiol Renal Physiol. 2010;299(2):F336–46.CrossRefPubMed Yang Z, Xiaohua W, et al. Uric acid increases fibronectin synthesis through upregulation of lysyl oxidase expression in rat renal tubular epithelial cells. Am J Physiol Renal Physiol. 2010;299(2):F336–46.CrossRefPubMed
23.
go back to reference Talbott JH, Terplan KL. The kidney in gout. Medicine (Baltimore). 1960;39:405–67.CrossRef Talbott JH, Terplan KL. The kidney in gout. Medicine (Baltimore). 1960;39:405–67.CrossRef
24.
go back to reference Syrjanen J, Mustonen J, et al. Hypertriglyceridaemia and hyperuricaemia are risk factors for progression of IgA nephropathy. Nephrol Dial Transplant. 2000;15(1):34–42.CrossRefPubMed Syrjanen J, Mustonen J, et al. Hypertriglyceridaemia and hyperuricaemia are risk factors for progression of IgA nephropathy. Nephrol Dial Transplant. 2000;15(1):34–42.CrossRefPubMed
25.
go back to reference Ohno I, Hosoya T, et al. Serum uric acid and renal prognosis in patients with IgA nephropathy. Nephron. 2001;87(4):333–9.CrossRefPubMed Ohno I, Hosoya T, et al. Serum uric acid and renal prognosis in patients with IgA nephropathy. Nephron. 2001;87(4):333–9.CrossRefPubMed
26.
go back to reference Hebert LA, Wilmer WA, et al. Renoprotection: one or many therapies? Kidney Int. 2001;59(4):1211–26.CrossRefPubMed Hebert LA, Wilmer WA, et al. Renoprotection: one or many therapies? Kidney Int. 2001;59(4):1211–26.CrossRefPubMed
27.
go back to reference Iseki K, Oshiro S, et al. Significance of hyperuricemia on the early detection of renal failure in a cohort of screened subjects. Hypertens Res. 2001;24(6):691–7.CrossRefPubMed Iseki K, Oshiro S, et al. Significance of hyperuricemia on the early detection of renal failure in a cohort of screened subjects. Hypertens Res. 2001;24(6):691–7.CrossRefPubMed
28.
go back to reference Kono H, Chen CJ, et al. Uric acid promotes an acute inflammatory response to sterile cell death in mice. J Clin Invest. 2010;210(6):1939–49.CrossRef Kono H, Chen CJ, et al. Uric acid promotes an acute inflammatory response to sterile cell death in mice. J Clin Invest. 2010;210(6):1939–49.CrossRef
29.
go back to reference Asada N, Takase M, et al. Dysfunction of fibroblasts of extrarenal origin underlies renal fibrosis and renal anemia in mice. J Clin Invest. 2011;121(10):3981–90.CrossRefPubMedPubMedCentral Asada N, Takase M, et al. Dysfunction of fibroblasts of extrarenal origin underlies renal fibrosis and renal anemia in mice. J Clin Invest. 2011;121(10):3981–90.CrossRefPubMedPubMedCentral
30.
go back to reference Humphreys BD, Lin SL, et al. Fate tracing reveals the pericyte and not epithelial origin of myofibroblasts in kidney fibrosis. Am J Pathol. 2010;176(1):85–97.CrossRefPubMedPubMedCentral Humphreys BD, Lin SL, et al. Fate tracing reveals the pericyte and not epithelial origin of myofibroblasts in kidney fibrosis. Am J Pathol. 2010;176(1):85–97.CrossRefPubMedPubMedCentral
31.
go back to reference Roch-Ramel F, Guisan B. Renal transport of Urate in humans. News Physiol Sci. 1999;14:80–4.PubMed Roch-Ramel F, Guisan B. Renal transport of Urate in humans. News Physiol Sci. 1999;14:80–4.PubMed
32.
go back to reference Corry DB, Eslami P, et al. Uric acid stimulates vascular smooth muscle cell proliferation and oxidative stress via the vascular renin-angiotensin system. J Hypertens. 2008;26(2):269–75.CrossRefPubMed Corry DB, Eslami P, et al. Uric acid stimulates vascular smooth muscle cell proliferation and oxidative stress via the vascular renin-angiotensin system. J Hypertens. 2008;26(2):269–75.CrossRefPubMed
34.
go back to reference Ryu ES, Kim MJ, et al. Uric acid-induced phenotypic transition of renal tubular cells as a novel mechanism of chronic kidney disease. Am J Physiol Renal Physiol. 2012;304(5):F471–80.CrossRef Ryu ES, Kim MJ, et al. Uric acid-induced phenotypic transition of renal tubular cells as a novel mechanism of chronic kidney disease. Am J Physiol Renal Physiol. 2012;304(5):F471–80.CrossRef
36.
go back to reference Lin SL, Kisseleva T, et al. Pericytes and perivascular fibroblasts are the primary source of collagen-producing cells in obstructive fibrosis of the kidney. Am J Pathol. 2008;173(6):1617–27.CrossRefPubMedPubMedCentral Lin SL, Kisseleva T, et al. Pericytes and perivascular fibroblasts are the primary source of collagen-producing cells in obstructive fibrosis of the kidney. Am J Pathol. 2008;173(6):1617–27.CrossRefPubMedPubMedCentral
37.
go back to reference Grgic I, Duffield JS, et al. The origin of interstitial myofibroblasts in chronic kidney disease. Pediatr Nephrol. 2012;27(2):183–93.CrossRefPubMed Grgic I, Duffield JS, et al. The origin of interstitial myofibroblasts in chronic kidney disease. Pediatr Nephrol. 2012;27(2):183–93.CrossRefPubMed
38.
go back to reference Chao H, Liu J, et al. Uric acid stimulates endothelin-1 gene expression associated with NADPH oxidase in human aortic smooth muscle cells. Acta Pharmacol Sin. 2008;29(11):1301–12.CrossRefPubMed Chao H, Liu J, et al. Uric acid stimulates endothelin-1 gene expression associated with NADPH oxidase in human aortic smooth muscle cells. Acta Pharmacol Sin. 2008;29(11):1301–12.CrossRefPubMed
39.
go back to reference Hocher B, Thone-Reineke C, et al. Endothelin-1 transgenic mice develop glomerulosclerosis, interstitial fibrosis, and renal cysts but not hypertension. J Clin Invest. 1997;99(6):1380–9.CrossRefPubMedPubMedCentral Hocher B, Thone-Reineke C, et al. Endothelin-1 transgenic mice develop glomerulosclerosis, interstitial fibrosis, and renal cysts but not hypertension. J Clin Invest. 1997;99(6):1380–9.CrossRefPubMedPubMedCentral
40.
41.
go back to reference Knerr I, Nyul Z, et al. Increased endothelin-1 and decreased adrenomedullin gene expression in the stenotic tissue of congenital pelvi-ureteric junction obstruction in children. BJU Int. 2001;87(7):667–71.CrossRefPubMed Knerr I, Nyul Z, et al. Increased endothelin-1 and decreased adrenomedullin gene expression in the stenotic tissue of congenital pelvi-ureteric junction obstruction in children. BJU Int. 2001;87(7):667–71.CrossRefPubMed
42.
go back to reference Goddard J, Eckhart C, et al. Endothelin a receptor antagonism and angiotensin-converting enzyme inhibition are synergistic via an endothelin B receptor-mediated and nitric oxide-dependent mechanism. J Am Soc Nephrol. 2004;15(10):2601–10.CrossRefPubMed Goddard J, Eckhart C, et al. Endothelin a receptor antagonism and angiotensin-converting enzyme inhibition are synergistic via an endothelin B receptor-mediated and nitric oxide-dependent mechanism. J Am Soc Nephrol. 2004;15(10):2601–10.CrossRefPubMed
43.
go back to reference Goddard J, Johnston NR, et al. Endothelin-a receptor antagonism reduces blood pressure and increases renal blood flow in hypertensive patients with chronic renal failure: a comparison of selective and combined endothelin receptor blockade. Circulation. 2004;109(9):1186–93.CrossRefPubMed Goddard J, Johnston NR, et al. Endothelin-a receptor antagonism reduces blood pressure and increases renal blood flow in hypertensive patients with chronic renal failure: a comparison of selective and combined endothelin receptor blockade. Circulation. 2004;109(9):1186–93.CrossRefPubMed
44.
go back to reference Arfian N, Emoto N, et al. ET-1 deletion from endothelial cells protects the kidney during the extension phase of ischemia/reperfusion injury. Biochem Biophys Res Commun. 2012;425(2):443–9.CrossRefPubMed Arfian N, Emoto N, et al. ET-1 deletion from endothelial cells protects the kidney during the extension phase of ischemia/reperfusion injury. Biochem Biophys Res Commun. 2012;425(2):443–9.CrossRefPubMed
Metadata
Title
Uric acid causes kidney injury through inducing fibroblast expansion, Endothelin-1 expression, and inflammation
Authors
Muhammad Mansyur Romi
Nur Arfian
Untung Tranggono
Wiwit Ananda Wahyu Setyaningsih
Dwi Cahyani Ratna Sari
Publication date
01-12-2017
Publisher
BioMed Central
Published in
BMC Nephrology / Issue 1/2017
Electronic ISSN: 1471-2369
DOI
https://doi.org/10.1186/s12882-017-0736-x

Other articles of this Issue 1/2017

BMC Nephrology 1/2017 Go to the issue