Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Review Article
  • Published:

Mouse models for human hyperuricaemia: a critical review

Abstract

Hyperuricaemia (increased serum urate concentration) occurs mainly in higher primates, including in humans, because of inactivation of the gene encoding uricase during primate evolution. Individuals with hyperuricaemia might develop gout — a painful inflammatory arthritis caused by monosodium urate crystal deposition in articular structures. Hyperuricaemia is also associated with common chronic diseases, including hypertension, chronic kidney disease, type 2 diabetes and cardiovascular disease. Many mouse models have been developed to investigate the causal mechanisms for hyperuricaemia. These models are highly diverse and can be divided into two broad categories: mice with genetic modifications (genetically induced models) and mice exposed to certain environmental factors (environmentally induced models; for example, pharmaceutical or dietary induction). This Review provides an overview of the mouse models of hyperuricaemia and the relevance of these models to human hyperuricaemia, with an emphasis on those models generated through genetic modifications. The challenges in developing and comparing mouse models of hyperuricaemia and future research directions are also outlined.

Key points

  • Hyperuricaemia occurs mainly in higher primates, including in humans, primarily owing to inactivation of the uricase gene during primate evolution, which resulted in subsequent evolution of human-specific physiology to tolerate this inactivation.

  • Mouse models of hyperuricaemia have been widely used to provide valuable insights into urate biology but do not yet reliably and consistently simulate the urate-mediated hyperuricaemia that occurs in humans.

  • Such models are potentially valuable resources for dissecting the mechanisms underlying hyperuricaemia as well as the progression from hyperuricaemia to gout and associated comorbidities.

  • A key challenge is to develop uricase-disabled model mice that can survive with increased urate levels and remain healthy and fertile.

  • Community-wide efforts are needed to reach consensus about the definition of hyperuricaemia in mice, to develop protocols for generating suitable models of hyperuricaemia and to adhere to a standard protocol for urate measurements.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: Urate metabolism in mammals.
Fig. 2: Generation of mouse models of hyperuricaemia.

Similar content being viewed by others

References

  1. Dalbeth, N., Merriman, T. R. & Stamp, L. K. Gout. Lancet 388, 2039–2052 (2016).

    CAS  Google Scholar 

  2. Ichida, K. et al. Decreased extra-renal urate excretion is a common cause of hyperuricemia. Nat. Commun. 3, 764 (2012). The investigators in this study generate Abcg2 -knockout mice that have increased urate concentrations and reduced gut excretion of urate. The renal excretion of urate is increased in these mice owing to overload of renal urate-handling machinery.

    PubMed  PubMed Central  Google Scholar 

  3. Dalbeth N. et al. Relationship between serum urate concentration and clinically evidence incident gout: an individual participant data analysis. Ann. Rheum. Dis. 77, 1048–1052 (2018).

    CAS  PubMed  Google Scholar 

  4. Bhole, V., Choi, J. W., Kim, S. W., de Vera, M. & Choi, H. Serum uric acid levels and the risk of type 2 diabetes: a prospective study. Am. J. Med. 123, 957–961 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  5. Borghi, C. et al. Serum uric acid and the risk of cardiovascular and renal disease. J. Hypertension 33, 1729–1741 (2015).

    CAS  Google Scholar 

  6. Kuo, C. F., Grainge, M. J., Zhang, W. & Doherty, M. Global epidemiology of gout: prevalence, incidence and risk factors. Nat. Rev. Rheumatol. 11, 649–662 (2015).

    PubMed  Google Scholar 

  7. Zhu, Y., Pandya, B. J. & Choi, H. K. Prevalence of gout and hyperuricemia in the US general population: the National Health and Nutrition Examination Survey 2007–2008. Arthritis Rheum. 63, 3136–3141 (2011).

    PubMed  Google Scholar 

  8. Miao, Z. et al. Dietary and lifestyle changes associated with high prevalence of hyperuricemia and gout in the Shandong coastal cities of Eastern China. J. Rheumatol. 35, 1859–1864 (2008).

    PubMed  Google Scholar 

  9. Robinson, P. C., Taylor, W. J. & Merriman, T. R. Systematic review of the prevalence of gout and hyperuricaemia in Australia. Intern. Med. J. 42, 997–1007 (2012).

    CAS  PubMed  Google Scholar 

  10. Chen-Xu, M., Yokose, C., Rai, S. K., Pillinger, M. H. & Choi, H. K. Contemporary prevalence of gout and hyperuricemia in the United States and decadal trends: the National Health and Nutrition Examination Survey 2007–2016. Arthritis Rheumatol. https://doi.org/10.1002/art.40807 (2019).

    Article  PubMed  Google Scholar 

  11. Alvarez-Lario, B. & Macarron-Vicente, J. Uric acid and evolution. Rheumatology 49, 2010–2015 (2010).

    CAS  PubMed  Google Scholar 

  12. Friedman, T. B., Polanco, G. E., Appold, J. C. & Mayle, J. E. On the loss of uricolytic activity during primate evolution—I. Silencing of urate oxidase in a hominoid ancestor. Comp. Biochem. Physiol. B 81, 653–659 (1985).

    CAS  PubMed  Google Scholar 

  13. Oda, M., Satta, Y., Takenaka, O. & Takahata, N. Loss of urate oxidase activity in hominoids and its evolutionary implications. Mol. Biol. Evol. 19, 640–653 (2002).

    CAS  PubMed  Google Scholar 

  14. Wu, X. et al. Hyperuricemia and urate nephropathy in urate oxidase-deficient mice. Proc. Natl Acad. Sci. USA 91, 742–746 (1994). This article is the first report of a uricase - knockout mouse, which was developed using ESC-targeting technology. The mice have increased urate concentrations and are viable and fertile, but the mice also have a high mortality owing to severe nephropathy.

    CAS  PubMed  Google Scholar 

  15. Ames, B. N., Cathcart, R., Schwiers, E. & Hochstein, P. Uric acid provides an antioxidant defense in humans against oxidant- and radical-caused aging and cancer: a hypothesis. Proc. Natl Acad. Sci. USA 78, 6858–6862 (1981).

    CAS  PubMed  Google Scholar 

  16. Chen, X. et al. Disrupted and transgenic urate oxidase alter urate and dopaminergic neurodegeneration. Proc. Natl Acad. Sci. USA 110, 300–305 (2013).

    CAS  PubMed  Google Scholar 

  17. Major, T. J., Dalbeth, N., Stahl, E. A. & Merriman, T. R. An update on the genetics of hyperuricaemia and gout. Nat. Rev. Rheumatol. 14, 341–353 (2018).

    CAS  PubMed  Google Scholar 

  18. Merriman, T. R. An update on the genetic architecture of hyperuricemia and gout. Arthritis Res. Ther. 17, 98 (2015).

    PubMed  PubMed Central  Google Scholar 

  19. Kanai, M. et al. Genetic analysis of quantitative traits in the Japanese population links cell types to complex human diseases. Nat. Genet. 50, 390–400 (2018).

    CAS  PubMed  Google Scholar 

  20. Köttgen, A. et al. Genome-wide association analyses identify 18 new loci associated with serum urate concentrations. Nat. Genet. 45, 145–154 (2013). This seminal GWAS identifies ~30 loci that are reproducibly associated with serum urate concentration in European individuals.

    PubMed  Google Scholar 

  21. Nakatochi, M. et al. Genome-wide meta-analysis identifies multiple novel loci associated with serum uric acid levels in Japanese individuals. Commun. Biol. 2, 115 (2019).

    Google Scholar 

  22. Cleophas, M. C. et al. ABCG2 polymorphisms in gout: insights into disease susceptibility and treatment approaches. Pharmacogenomics Pers. Med. 10, 129–142 (2017).

    CAS  Google Scholar 

  23. Ketharnathan, S. et al. A non-coding genetic variant maximally associated with serum urate levels is functionally linked to HNF4A-dependent PDZK1 expression. Hum. Mol. Genet. 27, 3964–3973 (2018).

    CAS  PubMed  Google Scholar 

  24. Chhana, A., Lee, G. & Dalbeth, N. Factors influencing the crystallization of monosodium urate: a systematic literature review. BMC Musculoskelet. Disord. 16, 296 (2015).

    PubMed  PubMed Central  Google Scholar 

  25. Lu, J. et al. Knockout of the urate oxidase gene provides a stable mouse model of hyperuricemia associated with metabolic disorders. Kidney Int. 93, 69–80 (2018). This study develops a uricase-deficient mouse line. The mice have increased serum urate concentrations, which are associated with the development of metabolic comorbidities, suggesting that these mice could be used as a model of hyperuricaemia-associated metabolic syndrome.

    CAS  PubMed  Google Scholar 

  26. Cook, S. A. et al. Mouse paracentric inversion In(3)55Rk mutates the urate oxidase gene. Cytogenet. Cell Genet. 93, 77–82 (2001).

    CAS  PubMed  Google Scholar 

  27. Vitart, V. et al. SLC2A9 is a newly identified urate transporter influencing serum urate concentration, urate excretion and gout. Nat. Genet. 40, 437–442 (2008).

    CAS  PubMed  Google Scholar 

  28. Caulfield, M. J. et al. SLC2A9 is a high-capacity urate transporter in humans. PLOS Med. 5, e197 (2008).

    PubMed  PubMed Central  Google Scholar 

  29. Preitner, F. et al. Glut9 is a major regulator of urate homeostasis and its genetic inactivation induces hyperuricosuria and urate nephropathy. Proc. Natl Acad. Sci. USA 106, 15501–15506 (2009). The investigators in this study generate Slc2a9 -knockout mice that have moderately increased urate concentrations. Unexpectedly, the mice do not develop urate nephropathy or structural abnormalities in the kidneys.

    CAS  PubMed  Google Scholar 

  30. Preitner, F. et al. Urate-induced acute renal failure and chronic inflammation in liver-specific Glut9 knockout mice. Am. J. Physiol. Renal Physiol. 305, F786–F795 (2013).

    CAS  PubMed  Google Scholar 

  31. Preitner, F. et al. No development of hypertension in the hyperuricemic liver-Glut9 knockout mouse. Kidney Int. 87, 940–947 (2015).

    CAS  PubMed  Google Scholar 

  32. DeBosch, B. J., Kluth, O., Fujiwara, H., Schurmann, A. & Moley, K. Early-onset metabolic syndrome in mice lacking the intestinal uric acid transporter SLC2A9. Nat. Commun. 5, 4642 (2014). The researchers of this study develop gut-enterocyte-specific Slc2a9 -knockout mice that have moderately increased urate concentrations and metabolic syndrome (including spontaneous hypertension, dyslipidaemia and elevated body fat). These mice might be suitable as a model of hyperuricaemia-associated metabolic syndrome.

    CAS  PubMed  PubMed Central  Google Scholar 

  33. Auberson, M. et al. SLC2A9 (GLUT9) mediates urate reabsorption in the mouse kidney. Pflugers Archiv. 470, 1739–1751 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  34. Dinour, D. et al. Homozygous mutations cause severe renal hypouricemia. J. Am. Soc. Nephrol. 21, 64–72 (2010).

  35. Johnson, R. J. et al. Hyperuricemia, acute and chronic kidney disease, hypertension and cardiovascular disease: report of a scientific workshop organized by the National Kidney Foundation. Am. J. Kidney Dis. 71, 851–865 (2018).

    PubMed  Google Scholar 

  36. Johnson, R. J., Merriman, T. & Lanaspa, M. A. Causal or noncausal relationship of uric acid with diabetes. Diabetes 64, 2720–2722 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  37. Robinson, P. C., Choi, H. K., Do, R. & Merriman, T. R. Insight into rheumatological cause and effect through the use of Mendelian randomization. Nat. Rev. Rheumatol. 12, 486–496 (2016).

    PubMed  Google Scholar 

  38. Li, X. et al. Serum uric acid levels and multiple health outcomes: umbrella review of evidence from observational studies, randomised controlled trials, and Mendelian randomisation studies. BMJ 357, j2376 (2017).

    PubMed  PubMed Central  Google Scholar 

  39. Woodward, O. M. et al. Identification of a urate transporter, ABCG2, with a common functional polymorphism causing gout. Proc. Natl Acad. Sci. USA 106, 10338–10342 (2009).

    CAS  PubMed  Google Scholar 

  40. Matsuo, H. et al. Common defects of ABCG2, a high-capacity urate exporter, cause gout: a function-based genetic analysis in a Japanese population. Sci. Transl Med. 1, 5ra11 (2009).

    PubMed  Google Scholar 

  41. Takada, T. et al. ABCG2 dysfunction increases serum uric acid by decreased intestinal urate excretion. Nucleosides Nucleotides Nucleic Acids 33, 275–281 (2014).

    CAS  PubMed  Google Scholar 

  42. Fridovich, I. The competitive inhibition of uricase by oxonate and by related derivatives of s-Triazines. J. Biol. Chem. 240, 2491–2494 (1965).

    CAS  PubMed  Google Scholar 

  43. Matsuo, H. et al. ABCG2 dysfunction causes hyperuricemia due to both renal urate underexcretion and renal urate overload. Sci. Rep. 4, 3755 (2014).

    PubMed  PubMed Central  Google Scholar 

  44. Kannangara, D. R. et al. Hyperuricaemia: contributions of urate transporter ABCG2 and the fractional renal clearance of urate. Ann. Rheum. Dis. 75, 1363–1365 (2015).

    PubMed  Google Scholar 

  45. Hoque, K. M. & Woodward, O. M. New mouse model of gout risk variant, ABCG2 Q141K, reveals unexpectedly severe molecular and functional defect in ABCG2 mediated intestinal uric acid secretion [abstract 2898]. Arthritis Rheumatol. 69 (Suppl. 10), 4187 (2017).

    Google Scholar 

  46. Sulem, P. et al. Identification of low-frequency variants associated with gout and serum uric acid levels. Nat. Genet. 43, 1127–1130 (2011).

    CAS  PubMed  Google Scholar 

  47. Tanner, C. et al. Population-specific resequencing associates the ATP-binding cassette subfamily C member 4 gene with gout in New Zealand Maori and Pacific men. Arthritis Rheumatol. 69, 1461–1469 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  48. Eraly, S. A. et al. Multiple organic anion transporters contribute to net renal excretion of uric acid. Physiol. Genomics 33, 180–192 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  49. Hosoyamada, M. et al. Establishment and analysis of SLC22A12 (URAT1) knockout mouse. Nucleosides Nucleotides Nucleic Acids 29, 314–320 (2010). In this study, Slc22a12 -knockout mice are generated that have considerably increased serum concentrations of urate, although the fractional excretion of urate in the knockout mice is higher than that of wild-type mice.

    CAS  PubMed  Google Scholar 

  50. Hosoyamada, M. et al. Urat1-Uox double knockout mice are experimental animal models of renal hypouricemia and exercise-induced acute kidney injury. Nucleosides Nucleotides Nucleic Acids 35, 543–549 (2016).

    CAS  PubMed  Google Scholar 

  51. Kocher, O., Pal, R., Roberts, M., Cirovic, C. & Gilchrist, A. Targeted disruption of the PDZK1 gene by homologous recombination. Mol. Cell. Biol. 23, 1175–1180 (2003).

    CAS  PubMed  PubMed Central  Google Scholar 

  52. Hillesheim, J. et al. Down regulation of small intestinal ion transport in PDZK1- (CAP70/NHERF3) deficient mice. Pflugers Arch. 454, 575–586 (2007).

    CAS  PubMed  Google Scholar 

  53. Charkoftaki, G. et al. Transcriptomic analysis and plasma metabolomics in Aldh16a1-null mice reveals a potential role of ALDH16A1 in renal function. Chem. Biol. Interact. 276, 15–22 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  54. Lin, Z. P. et al. Disruption of cAMP and prostaglandin E2 transport by multidrug resistance protein 4 deficiency alters cAMP-mediated signaling and nociceptive response. Mol. Pharmacol. 73, 243–251 (2008).

    CAS  PubMed  Google Scholar 

  55. Dankers, A. C. A. et al. Hyperuricemia influences tryptophan metabolism via inhibition of multidrug resistance protein 4 (MRP4) and breast cancer resistance protein (BCRP). Biochim. Biophys. Acta 1832, 1715–1722 (2013).

    CAS  PubMed  Google Scholar 

  56. Aziz, A., Soucie, E., Sarrazin, S. & Sieweke, M. H. MafB/c-Maf deficiency enables self-renewal of differentiated functional macrophages. Science 326, 867–871 (2009).

    CAS  PubMed  Google Scholar 

  57. Katsuoka, F., Yamazaki, H. & Yamamoto, M. Small Maf deficiency recapitulates the liver phenotypes of Nrf1- and Nrf2-deficient mice. Genes Cells 21, 1309–1319 (2016).

    CAS  PubMed  Google Scholar 

  58. Lyon, M. F. et al. A dominant mutation within the DNA-binding domain of the bZIP transcription factor Maf causes murine cataract and results in selective alteration in DNA binding. Hum. Mol. Genet. 12, 585–594 (2003).

    CAS  PubMed  Google Scholar 

  59. Kelley, W. N., Rosenbloom, F. M., Henderson, J. F. & Seegmiller, J. E. A specific enzyme defect in gout associated with overproduction of uric acid. Proc. Natl Acad. Sci. USA 57, 1735–1739 (1967).

    CAS  PubMed  Google Scholar 

  60. Zennaro, C. et al. The renal phenotype of allopurinol-treated HPRT-deficient mouse. PLOS ONE 12, e0173512 (2017).

    PubMed  PubMed Central  Google Scholar 

  61. 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).

    PubMed  PubMed Central  Google Scholar 

  62. Stevenson, W. S. et al. Deficiency of 5-hydroxyisourate hydrolase causes hepatomegaly and hepatocellular carcinoma in mice. Proc. Natl Acad. Sci. USA 107, 16625–16630 (2010).

    CAS  PubMed  Google Scholar 

  63. Baey, C., Yang, J., Ronchese, F. & Harper, J. L. Hyperuricaemic Urah Plt2/Plt2 mice show altered T cell proliferation and defective tumor immunity after local immunotherapy with Poly I:C. PLOS ONE 13, e0206827 (2018).

    PubMed  PubMed Central  Google Scholar 

  64. Wu, X. H. et al. Riparoside B and timosaponin J, two steroidal glycosides from Smilax riparia, resist to hyperuricemia based on URAT1 in hyperuricemic mice. Phytomedicine 21, 1196–1201 (2014).

    CAS  PubMed  Google Scholar 

  65. Wang, M. X., Liu, Y. L., Yang, Y., Zhang, D. M. & Kong, L. D. Nuciferine restores potassium oxonate-induced hyperuricemia and kidney inflammation in mice. Eur. J. Pharmacol. 747, 59–70 (2015).

    CAS  PubMed  Google Scholar 

  66. Wang, M., Zhao, J., Zhang, N. & Chen, J. Astilbin improves potassium oxonate-induced hyperuricemia and kidney injury through regulating oxidative stress and inflammation response in mice. Biomed. Pharmacother. 83, 975–988 (2016).

    CAS  PubMed  Google Scholar 

  67. Chen, G., Tan, M. L., Li, K. K., Leung, P. C. & Ko, C. H. Green tea polyphenols decreases uric acid level through xanthine oxidase and renal urate transporters in hyperuricemic mice. J. Ethnopharmacol. 175, 14–20 (2015).

    CAS  PubMed  Google Scholar 

  68. Watanabe, T., Tomioka, N. H., Watanabe, S., Tsuchiya, M. & Hosoyamada, M. False in vitro and in vivo elevations of uric acid levels in mouse blood. Nucleosides Nucleotides Nucleic Acids 33, 192–198 (2014). This study highlights the extreme variability in mouse urate concentrations measured across different studies. There is a 19-fold difference in urate concentrations between live and dead mice sampled for blood urate measurements.

    CAS  PubMed  Google Scholar 

  69. Li, J. M., Zhang, X., Wang, X., Xie, Y. C. & Kong, L. D. Protective effects of cortex fraxini coumarines against oxonate-induced hyperuricemia and renal dysfunction in mice. Eur. J. Pharmacol. 666, 196–204 (2011).

    CAS  PubMed  Google Scholar 

  70. Hu, Q. H., Zhang, X., Wang, X., Jiao, R. Q. & Kong, L. D. Quercetin regulates organic ion transporter and uromodulin expression and improves renal function in hyperuricemic mice. Eur. J. Nutr. 51, 593–606 (2012).

    CAS  PubMed  Google Scholar 

  71. Liu, Y. L. et al. Betaine reduces serum uric acid levels and improves kidney function in hyperuricemic mice. Planta Med. 80, 39–47 (2014).

    CAS  PubMed  Google Scholar 

  72. Yong, T. et al. Actions of water extract from Cordyceps militaris in hyperuricemic mice induced by potassium oxonate combined with hypoxanthine. J. Ethnopharmacol. 194, 403–411 (2016).

    PubMed  Google Scholar 

  73. Sun, W. F. et al. MicroRNA expression patterns of the kidney in hyperuricemia mice treated with Xiezhuo Chubi Decoction. Chin. J. Integr. Med. 17, 35–42 (2011).

    PubMed  Google Scholar 

  74. Xu, C. et al. Xanthine oxidase in non-alcoholic fatty liver disease and hyperuricemia: one stone hits two birds. J. Hepatol. 62, 1412–1419 (2015).

    CAS  PubMed  Google Scholar 

  75. Nakatsu, Y. et al. The xanthine oxidase inhibitor febuxostat suppresses development of nonalcoholic steatohepatitis in a rodent model. Am. J. Physiol. Gastrointest. Liver Physiol. 309, G42–G51 (2015).

    CAS  PubMed  Google Scholar 

  76. Bakker, P. J. et al. Nlrp3 is a key modulator of diet-induced nephropathy and renal cholesterol accumulation. Kidney Int. 85, 1112–1122 (2014).

    CAS  PubMed  Google Scholar 

  77. Mutel, E. et al. Targeted deletion of liver glucose-6 phosphatase mimics glycogen storage disease type 1a including development of multiple adenomas. J. Hepatol. 54, 529–537 (2011).

    CAS  PubMed  Google Scholar 

  78. Ito, T. et al. Glomerular changes in the KK-Ay/Ta mouse: a possible model for human type 2 diabetic nephropathy. Nephrology 11, 29–35 (2006).

    CAS  PubMed  Google Scholar 

  79. Adachi, S. I., Yoshizawa, F. & Yagasaki, K. Hyperuricemia in type 2 diabetic model KK-Ay/Ta mice: a potent animal model with positive correlation between insulin resistance and plasma high uric acid levels. BMC Res. Notes 10, 577 (2017).

    PubMed  PubMed Central  Google Scholar 

  80. Baldwin, W. et al. Hyperuricemia as a mediator of the proinflammatory endocrine imbalance in the adipose tissue in a murine model of the metabolic syndrome. Diabetes 60, 1258–1269 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  81. Shaw, O. M., Pool, B., Dalbeth, N. & Harper, J. L. The effect of diet-induced obesity on the inflammatory phenotype of non-adipose-resident macrophages in an in vivo model of gout. Rheumatology 53, 1901–1905 (2014).

    CAS  PubMed  Google Scholar 

  82. Marchetti, C. et al. NLRP3 inflammasome inhibitor OLT1177 suppresses joint inflammation in murine models of acute arthritis. Arthritis Res. Ther. 20, 169 (2018).

    PubMed  PubMed Central  Google Scholar 

  83. Amaral, F. A. et al. Transmembrane TNF-α is sufficient for articular inflammation and hypernociception in a mouse model of gout. Eur. J. Immunol. 46, 204–211 (2016).

    CAS  PubMed  Google Scholar 

  84. Liu-Bryan, R., Scott, P., Sydlaske, A., Rose, D. M. & Terkeltaub, R. Innate immunity conferred by Toll-like receptors 2 and 4 and myeloid differentiation factor 88 expression is pivotal to monosodium urate monohydrate crystal-induced inflammation. Arthritis Rheum. 52, 2936–2946 (2005).

    CAS  PubMed  Google Scholar 

  85. Lee, Y. M., Shon, E. J., Kim, O. S. & Kim, D. S. Effects of Mollugo pentaphylla extract on monosodium urate crystal-induced gouty arthritis in mice. BMC Complement. Altern. Med. 17, 447 (2017).

    PubMed  PubMed Central  Google Scholar 

  86. Cumpelik, A., Ankli, B., Zecher, D. & Schifferli, J. A. Neutrophil microvesicles resolve gout by inhibiting C5a-mediated priming of the inflammasome. Ann. Rheum. Dis. 75, 1236–1245 (2016).

    CAS  PubMed  Google Scholar 

  87. Crisan, T. O. et al. Soluble uric acid primes TLR-induced proinflammatory cytokine production by human primary cells via inhibition of IL-1Ra. Ann. Rheum. Dis. 75, 755–762 (2016).

    CAS  PubMed  Google Scholar 

  88. Mazzali, M. et al. Elevated uric acid increases blood pressure in the rat by a novel crystal-independent mechanism. Hypertension 38, 1101–1106 (2001).

    CAS  PubMed  Google Scholar 

  89. Su, Q. et al. Hypouricemic and nephroprotective effects of an active fraction from Polyrhachis Vicina Roger on potassium oxonate-induced hyperuricemia in rats. Kidney Blood Press. Res. 43, 220–233 (2018).

    CAS  PubMed  Google Scholar 

  90. Liu, X., Chen, R., Shang, Y., Jiao, B. & Huang, C. Lithospermic acid as a novel xanthine oxidase inhibitor has anti-inflammatory and hypouricemic effects in rats. Chem. Biol. Interact. 176, 137–142 (2008).

    CAS  PubMed  Google Scholar 

  91. Leask, M. et al. Functional urate-associated genetic variants influence expression of lincRNAs LINC01229 and MAFTRR. Front. Genet. 9, 733 (2018).

    PubMed  Google Scholar 

  92. Zhang, Y., Li, Q., Wang, F. & Xing, C. A zebrafish (danio rerio) model for high-throughput screening food and drugs with uric acid-lowering activity. Biochem. Biophys. Res. Commun. 508, 494–498 (2019).

    CAS  PubMed  Google Scholar 

  93. Marchetti, M. et al. Catalysis and structure of zebrafish urate oxidase provide insights into the origin of hyperuricemia in hominoids. Sci. Rep. 6, 38302 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  94. Hall, C. J. et al. Blocking fatty acid-fueled mROS production within macrophages alleviates acute gouty inflammation. J. Clin. Invest. 128, 1752–1771 (2018).

    PubMed  PubMed Central  Google Scholar 

  95. Remy, C., Richert, D. A. & Westerfeld, W. W. The determination of xanthine dehydrogenase in chicken tissues. J. Biol. Chem. 192, 649–657 (1951).

    Google Scholar 

  96. Ejaz, S., Kim, B. S. & Lim, C. W. Gout induced by intoxication of sodium bicarbonate in Korean native broilers. Drug Chem. Toxicol. 28, 245–261 (2005).

    CAS  PubMed  Google Scholar 

  97. Poffers, J., Lumeij, J. T., Timmermans-Sprang, E. P. & Redig, P. T. Further studies on the use of allopurinol to reduce plasma uric acid concentrations in the Red-tailed Hawk (Buteo jamaicensis) hyperuricaemic model. Avian Pathol. 31, 567–572 (2002).

    CAS  PubMed  Google Scholar 

  98. Lin, Z., Zhang, B., Liu, X., Jin, R. & Zhu, W. Effects of chicory inulin on serum metabolites of uric acid, lipids, glucose, and abdominal fat deposition in quails induced by purine-rich diets. J. Med. Food 17, 1214–1221 (2014).

    CAS  PubMed  Google Scholar 

  99. Zhang, X. H. et al. Molecular characterization, balancing selection, and genomic organization of the tree shrew (Tupaia belangeri) MHC class I gene. Gene 522, 147–155 (2013).

    CAS  PubMed  Google Scholar 

  100. Tang, D. H. et al. Potassium oxonate induces acute hyperuricemia in the tree shrew (tupaia belangeri chinensis). Exp. Anim. 66, 209–216 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  101. Szczurek, P. et al. Oral uricase eliminates blood uric acid in the hyperuricemic pig model. PLOS ONE 12, e0179195 (2017).

    PubMed  PubMed Central  Google Scholar 

  102. Bannasch, D. et al. Mutations in the SLC2A9 gene cause hyperuricosuria and hyperuricemia in the dog. PLOS Genet. 4, e1000246 (2008).

    PubMed  PubMed Central  Google Scholar 

  103. Rothschild, B. M., Tanke, D. & Carpenter, K. Tyrannosaurs suffered from gout. Nature 387, 357 (1997).

    CAS  PubMed  Google Scholar 

  104. Ammerman, L. E. Resurrecting Tyrannosaurus rex. Thesis, Baylor Univ. Texas (2016).

  105. Fatima, T. et al. The relationship between ferritin and urate levels and risk of gout. Arthritis Res. Ther. 20, 179 (2018).

    PubMed  PubMed Central  Google Scholar 

  106. Topless, R. K. et al. Association of SLC2A9 genotype with phenotypic variability of serum urate in pre-menopausal women. Front. Genet. 6, 313 (2015).

    PubMed  PubMed Central  Google Scholar 

  107. 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).

    CAS  PubMed  PubMed Central  Google Scholar 

  108. Kratzer, J. T. et al. Evolutionary history and metabolic insights of ancient mammalian uricases. Proc. Natl Acad. Sci. USA 111, 3763–3768 (2014).

    CAS  PubMed  Google Scholar 

  109. Tan, P. K., Farrar, J. E., Gaucher, E. A. & Miner, J. N. Coevolution of URAT1 and uricase during primate evolution: implications for serum urate homeostasis and gout. Mol. Bio. Evol. 33, 2193–2200 (2016).

    CAS  Google Scholar 

  110. Kelly, S. J. et al. Diabetes insipidus in uricase-deficient mice: a model for evaluating therapy with poly(ethylene glycol)-modified uricase. J. Am. Soc. Nephrol. 12, 1001–1009 (2001).

    CAS  PubMed  Google Scholar 

  111. Zhang, F., Wen, Y. & Guo, X. CRISPR/Cas9 for genome editing: progress, implications and challenges. Hum. Mol. Genet. 23, R40–R46 (2014).

    CAS  PubMed  Google Scholar 

  112. Cleveland, B. M., Leonard, S. S., Klandorf, H. & Blemings, K. P. Urate oxidase knockdown decreases oxidative stress in a murine hepatic cell line. Oxid. Med. Cell. Longev. 2, 93–98 (2009).

    PubMed  PubMed Central  Google Scholar 

  113. Silva, A. J. et al. Mutant mice and neuroscience: recommendations concerning genetic background. Neuron 19, 755–759 (1997).

    Google Scholar 

  114. Montagutelli, X. Effect of the genetic background on the phenotype of mouse mutations. J. Am. Soc. Nephrol. 11, S101–S105 (2000).

    PubMed  Google Scholar 

  115. Dalbeth, N. et al. Relationship between serum urate concentration and clinically evident incident gout: an individual participant data analysis. Ann. Rheum. Dis. 77, 1048–1052 (2018).

    CAS  PubMed  Google Scholar 

  116. Terkeltaub, R. What makes gouty inflammation so variable? BMC Med. 15, 158 (2017).

    PubMed  PubMed Central  Google Scholar 

  117. Schauer, C. et al. Aggregated neutrophil extracellular traps limit inflammation by degrading cytokines and chemokines. Nat. Med. 20, 511–517 (2014).

    CAS  PubMed  Google Scholar 

  118. Pascual, E., Addadi, L., Andres, M. & Sivera, F. Mechanisms of crystal formation in gout-a structural approach. Nat. Rev. Rheumatol. 11, 725–730 (2015).

    CAS  PubMed  Google Scholar 

  119. Mandal, A. K. & Mount, D. B. The molecular physiology of uric acid homeostasis. Annu. Rev. Physiol. 77, 323–345 (2015).

    CAS  PubMed  Google Scholar 

  120. Sutin, A. R. et al. Impulsivity is associated with uric acid: evidence from humans and mice. Biol. Psychiatry 75, 31–37 (2014).

    CAS  PubMed  Google Scholar 

  121. Lu, J. et al. Urate-lowering therapy alleviates atherosclerosis inflammatory response factors and neointimal lesions in a mouse model of induced carotid atherosclerosis. FEBS J. 286, 1346–1359 (2019).

    CAS  PubMed  Google Scholar 

  122. Lizio, M. et al. Gateways to the FANTOM5 promoter level mammalian expression atlas. Genome Biol. 16, 22 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  123. RIKEN. Functional annotation of the mammalian genome. FANTOM http://fantom.gsc.riken.jp/ (2014).

  124. The Scripps Research Institute. BioGPS. BioGPS http://biogps.org (2019).

  125. Wu, C., Jin, X., Tsueng, G., Afrasiabi, C. & Su, A. I. BioGPS: building your own mash-up of gene annotations and expression profiles. Nucl. Acids Res. 44, D313–D316 (2016).

    CAS  PubMed  Google Scholar 

  126. Hruz, T. et al. Genevestigator V3: a reference expression database for the meta-analysis of transcriptomes. Adv. Bioinformatics 2008, 420747 (2008).

    PubMed  PubMed Central  Google Scholar 

  127. Nebion. Genevestigator. Geneinvestigator https://genevestigator.com/gv/ (2018).

Download references

Acknowledgements

The authors thank S. Robertson and the Clinical Genetics Group in the University of Otago, Dunedin, New Zealand, for their valuable discussions of this Review. J.L. is grateful for the support of the Departments of Women’s and Children’s Health, Biochemistry and Pathology, University of Otago. W.-H.W. is funded by Cure Kids NZ and the University of Otago.

Reviewer information

Nature Reviews Rheumatology thanks H. -K. Ea and F. Lioté for their contribution to the peer review of this work.

Author information

Authors and Affiliations

Authors

Contributions

J.L., C.L. and W.-H.W. researched data for the article. N.D., T.R.M. and W.-H.W. contributed to discussion of content. J.L., N.D., H.Y., T.R.M. and W.-H.W. contributed to writing the article, and all authors reviewed and edited the manuscript before submission.

Corresponding authors

Correspondence to Tony R. Merriman or Wen-Hua Wei.

Ethics declarations

Competing interests

N.D. declares that she has received research grant funding from Amgen and AstraZeneca; speaker fees from AbbVie, Janssen and Pfizer; and consulting fees from AstraZeneca, Horizon and Kowa. T.R.M. declares he has received research grant funding from Ardea Biosciences and Ironwood Pharmaceuticals and has received consulting fees from Ironwood Pharmaceuticals. The other authors declare that they have no competing interests.

Additional information

Publisher’s note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Glossary

Mendelian ratios

The expected ratios of genotypes at a locus observed in offspring under Mendel’s law of independent assortment; if one allele is embryonically lethal, the ratio will be skewed.

Mendelian randomization

The use of genetic variation in genes of known function to examine the causal effect of an exposure on disease in observational studies.

Pseudogenization

The process of generating a pseudogene, which is a gene that has DNA segments related to a real gene but has lost some or all functionality during evolution.

Congenic

A congenic mouse strain has a defined segment from a donor strain introduced into its genome.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Lu, J., Dalbeth, N., Yin, H. et al. Mouse models for human hyperuricaemia: a critical review. Nat Rev Rheumatol 15, 413–426 (2019). https://doi.org/10.1038/s41584-019-0222-x

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41584-019-0222-x

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing