7,546
Views
69
CrossRef citations to date
0
Altmetric
Review Article

Uric acid transporters hiding in the intestine

, , &
Pages 3151-3155 | Received 10 Jan 2016, Accepted 24 May 2016, Published online: 26 Aug 2016

Abstract

Context: Hyperuricaemia is known as an abnormally increased uric acid level in the blood. Although it was observed many years ago, since uric acid excretion via the intestine pathway accounted for approximately one-third of total elimination of uric acid, the molecular mechanism of ‘extra-renal excretion’ was poorly understood until the finding of uric acid transporters.

Objective: The objective of this study was to gather all information related to uric acid transporters in the intestine and present this information as a comprehensive and systematic review article.

Methods: A literature search was performed from various databases (e.g., Medline, Science Direct, Springer Link, etc.). The key terms included uric acid, transporter and intestine. The period for the search is from the 1950s to the present. The bibliographies of papers relating to the review subject were also searched for further relevant references.

Results: The uric acid transporters identified in the intestine are discussed in this review. The solute carrier (SLC) transporters include GLUT9, MCT9, NPT4, NPT homolog (NPT5) and OAT10. The ATP binding cassette (ABC) transporters include ABCG2 (BCRP), MRP2 and MRP4. Bacterial transporter YgfU is a low-affinity and high-capacity transporter for uric acid.

Conclusion: The present review may be helpful for further our understanding of hyperuricaemia and be of value in designing future studies on novel therapeutic pathways.

Introduction

Gout, a painful form of inflammatory arthritis, emerged as a ‘wealthy’ epidemic in the eighteenth century England. The epidemiological link between hyperuricaemia and gout was established nearly two centuries ago (Richette & Bardin Citation2010). An abnormally high level of blood uric acid is known as hyperuricaemia. With the formation of urate (the ionized form of uric acid) crystals, hyperuricaemia causes gout in the joints and predisposes to nephrolithiasis in the kidney. Recent evidence (Lima et al. Citation2015; Perez-Ruiz et al. Citation2015) is accumulating indicating a role for hyperuricaemia in the genesis and progression of hypertension, diabetes mellitus, metabolic syndrome, renal diseases, cardiovascular diseases and even the aging process (Campion et al. Citation1987; Iwama et al. Citation2012; Krishnan et al. Citation2014). In both academic and clinical practice, there has been increased interest in hyperuricaemia worldwide. Uric acid has now been identified as a predictor of incident events of these diseases (So & Thorens Citation2010).

The molecular physiology of uric acid homeostasis

Uric acid is produced by humans as the final metabolic product of purines, both endogenous and exogenous. Uric acid is a weak acid (pKa 5.8) and has low solubility in water as well as in plasma. At physiological pH, uric acid circulates mainly in the form of urate. Hyperuricaemia pathogenicity is associated with low solubility of urate in the extracellular environment leading to crystal formation. The levels of urate in the blood are maintained by the balance between uric acid generation and excretion (de Oliveira & Burini Citation2012). An alteration in this balance may account for hyperuricaemia, so either decreased excretion, increased synthesis or both will lead to elevated plasma uric acid concentrations (hyperuricaemia). Although dietary, genetic, or disease-related excesses in uric acid production underlie hyperuricaemia in some cases, practically the main cause for hyperuricaemia is lower excretion of uric acid. Among 65 hyperuricaemic patients analyzed for uric acid metabolism, 9.2% had over-production type, 80.0% had under-excretion type and 10.8% had a mixed type (Oka et al. Citation2014).

In the blood, urate would theoretically reach saturation in the concentration of 6.4 mg/dL, which may not occur because protein (mainly albumin) bound urate increases its solubility (Susic & Frohlich Citation2015). Similarly, the binding to protein is necessary for uric acid to be cleared from the circulation. Excretion of uric acid requires specialized transporters located in renal tubule cells and intestinal epithelial cells. The importance of the transporters in excretion of uric acid has become increasingly evident.

Uric acid transport by the kidney has been investigated for many years. The kidney plays a crucial role in maintaining plasma urate levels through complex transepithelial transport systems that promote both re-absorption and secretion of uric acid (Hediger et al. Citation2005). The primary transporter involved in uric acid re-absorption is urate transporter 1 (URAT1), which is only expressed in the kidney (Enomoto et al. Citation2002). Molecular identification of URAT1 promoted the advances of new transporters involved in renal uric acid transport. Many of transporters have been identified and well reviewed (Hediger et al. Citation2005; Richette & Bardin Citation2010; So & Thorens Citation2010). Recently, genome-wide association studies indicated a substantial association between uric acid concentration and single nucleotide polymorphisms (SNPs) of transporter-coding genes such as SLC2A9 (GLUT9), ABCG2 (BCRP), SLC17A1 (NPT1), SLC17A3 (NPT4), SLC17A4 (NPT5), SLC22A11 (OAT4), SLC22A12 (URAT1) and SLC16A9 (MCT9). GLUT9 (glucose transporter 9) has unexpectedly been shown to play a critical role in uric acid re-absorption at the proximal tubule, probably more prominent than its partner URAT1 (Sakurai Citation2013).

Uric acid transporters in the intestine

The intestine can also secrete uric acid. As far back as half a century ago, it was estimated that approximately one-third of uric acid was excreted into the gut and was further metabolized by resident bacteria (Buzard et al. Citation1952; Sorensen Citation1965). In oxonate-treated rats, significant amounts of externally administered and endogenous uric acid were recovered in the intestinal lumen, while biliary excretion was minimal. Accordingly, direct intestinal secretion was thought to be a substantial contributor to extra-renal elimination of uric acid (Hosomi et al. Citation2012). It is not difficult to imagine that the intestinal contribution of uric acid excretion is an important alternative pathway during renal insufficiency. Enhanced enteric excretion of uric acid may account for the lack of significant hyperuricaemia in chronic renal failure (Vaziri et al. Citation1995). In most gouty subjects, extra-renal disposal of uric acid is even greater. In some patients with renal insufficiency, enteral uricolysis constitutes the major process of uric acid disposal (Sorensen Citation1975). However, there had been few mechanistic studies on the intestinal ‘extra-renal excretion’ pathway of uric acid before the finding of UA transporters.

In the intestine, purine compounds are metabolized to uric acid, which is transported. The small intestine is a major site of uric acid production in the body (Oh et al. Citation1967). Furthermore, the small intestine represents the primary site of absorption for all ingested compounds. Intestinal transporters play significant roles in the absorbing process. P-Glycoprotein (P-gp), the first clinically important drug transporter, was identified in 1986 (Roninson et al. Citation1986).

An interest for other intestinal transporters is now emerging. The largest human intestinal transporter database (>5000 interaction entries for >3700 molecules) have been assembled and curated (Sedykh et al. Citation2013). Among those transporters mediating the cellular uptake and efflux of a broad variety of endogenous compounds, drugs and their metabolites, solute carriers (SLCs) and ATP binding cassette (ABC) transporters represent two superfamilies in humans. ABC transporters (e.g., multidrug resistance proteins, MRPs) utilize the energy from ATP hydrolysis and function as efflux transporters. SLC transporters (e.g., organic anion transporters, OATs) do not rely directly on ATP hydrolysis and are largely (but not exclusively) uptake importers moving small molecules from the extracellular milieu into cells (Estudante et al. Citation2013). While much research has been conducted focusing on the role of uric acid transporters in the kidney, less is known about the importance of uptake and efflux transporters identified in the intestine. The molecular nature of uric acid transporters in the intestine has so far not been investigated in detail.

ABCG2/BCRP

The ATP-binding cassette, subfamily G, 2 (ABCG2) was originally identified in placental tissue as a xenobiotic transporter in relation to the development of multidrug resistance of breast cancer cells. It was, therefore, also termed ‘breast cancer resistance protein’ (BCRP) (Doyle et al. Citation1998). ABCG2 is expressed on the apical membrane in tissues including kidney and intestine. The story of ABCG2 and uric acid began with a genome-wide association study, which indicated that ABCG2 played a crucial role of uric acid homeostasis through both renal and extra-renal uric acid excretion (Dehghan et al. Citation2008; Nakayama et al. Citation2011). At least 10% of all gout cases in whites are attributable to rs2231142, a common functional polymorphism in ABCG2 (Woodward et al. Citation2009). ABCG2 dysfunction caused renal uric acid under-excretion and induced hyperuricaemia even if the renal uric acid overload was not remarkable (Matsuo et al. Citation2014).

Recent studies suggest that ABCG2 has an important role in intestinal excretion. Given higher expression of ABCG2 in the intestine, it is likely that the hypoactive variant of ABCG2 leads to decreased uric acid excretion into the intestine rather than decreased uric acid excretion from the kidney (Sarkadi et al. Citation2004). In Caco-2 cells, uric acid showed a polarized flux from the basolateral to apical side, and this flux was almost abolished in the presence of an ABCG2 inhibitor (Hosomi et al. Citation2012). ABCG2 knockout mice significantly exhibited a reduction of intestinal uric acid excretion and an increased plasma concentration of uric acid (Ichida et al. Citation2012). In the 5/6 nephrectomy rat model of chronic kidney disease, serum UA did not increase despite the decrease in UA excretion. Meanwhile, ABCG2 was significantly increased in the intestine (Yano et al. Citation2014). Increased serum uric acid in patients with ABCG2 dysfunction could be explained by the decreased excretion of uric acid from the intestine (Takada et al. Citation2014). Patients with mild to severe ABCG2 dysfunction accounted for 88.2% of early-onset gout patients. Severe ABCG2 dysfunction particularly increased the risk of early-onset gout (Matsuo et al. Citation2013). These results suggest that decreased intestinal uric acid excretion caused by ABCG2 dysfunction might be a common mechanism of hyperuricemia.

Multidrug resistance proteins (MRPs, ABCCs)

MRP2 and MRP4 both have been shown to transport p-aminohippurate (PAH), the classical substrate used in the characterization of organic anion transport in renal proximal tubular cells. Various inhibitors of MRP2-mediated PAH transport also inhibited MRP4. However, MRP4 has higher affinity for PAH and is expressed more than MRP2 in the kidney (Smeets et al. Citation2004). With respect to renal physiology and pharmacology, particular attention should be paid to MRP2 and MRP4. In the apical membrane of kidney proximal tubules, MRP4 mediates ATP-dependent uric acid transport through multiple allosteric binding sites. MRP4 appears to control ATP dependent uric acid extrusion from the cells into the tubular lumen and thus contributes to uric acid excretion. Human MRP4, but not MRP2, expressed in HEK293 cells, mediates ATP-dependent efflux of uric acid (Van Aubel et al. Citation2004). Cultured avian proximal tubules lack re-absorptive uric acid transport, knockdown of MRP4 expression reduces uric acid secretion by 65% (Bataille et al. Citation2008).

MRPs forms 1 to 6 are present in intestinal epithelial cells (Grandvuinet et al. Citation2012). In jejunal tissue, MRP2 is highly expressed while MRP4 is expressed to a low extent (Hilgendorf et al. Citation2007). In cultured chicken proximal tubules, RT-PCR revealed mRNA for MRP2- and MRP4-like organic anion transporters in avian proximal epithelium. Luminal application of MK-571 (a known substrate for MRP2 and MRP4) dramatically reduced both uric acid secretory and re-absorptive flux (Dudas et al. Citation2005).

Glucose transporter 9 (GLUT9)

GLUT9 was initially identified by sequence similarity with the glucose transporter. Differing from other GLUTs, GLUT9 is most probably not involved in glucose and/or fructose transport (Mueckler & Thorens Citation2013). The relationship between GLUT9 and uric acid has recently been highlighted. Common variation in the SLC2A9 gene that encodes GLUT9 might explain 1.7–5.3% of the variance in serum uric acid concentrations, following a genome-wide association scan (Vitart et al. Citation2008). Despite the ever-increasing list of hyperuricaemic genes, variation in SLC2A9 remains the major single genetic determinant of serum uric acid, followed closely by the effects of variation in the ABCG2 gene (Mandal & Mount Citation2015). Mice with systemic knockout of Glut9 displayed moderate hyperuricaemia, massive hyperuricosuria and mild renal insufficiency (Preitner et al. Citation2009).

Human GLUT9 has two splice variants with different expression patterns: GLUT9a and GLUT9b. The two splice variants are expressed differentially within polarized cells, with GLUT9a localized predominantly on the basolateral surfaces and GLUT9b expressed on apical surfaces. The two isoforms could not be distinguished by the use of antibody, so the polarized distribution of the two splice variants is not confirmed in vivo (Augustin et al. Citation2004). GLUT9 is widely expressed, with substantial transcript levels primarily in kidney and liver, but is present at low levels in several other tissues, including small intestine and colon. GLUT9 is also expressed in articular cartilage, cytokines could induce expression of GLUT9 in chondrocytes, and thus it is possible that GLUT9 plays a role in gout (Phay et al. Citation2000). Because GLUT9 is present in the intestine, its genetic inactivation may lower intestinal excretion of uric acid, although this has not been formally tested. Enterocyte uric acid metabolism could potentially be targeted to modulate or prevent the development of the metabolic syndrome. Glut9-deficient mice developed impaired enterocyte uric acid transport kinetics, hyperuricaemia, hyperuricosuria, spontaneous hypertension, dyslipidaemia and elevated body fat (DeBosch et al. Citation2014).

Monocarboxylate transporter 9 (MCT9)

MCT9, a member of the monocarboxylate co-transporter family, was ubiquitously expressed and was especially expressed at a high level in the kidney (Halestrap & Price Citation2004). SLC16A9, encoding MCT9, was identified from analysis of the human genomic expressed sequence tag (EST) databases (Halestrap & Price Citation1999). Meta-analysis of genome-wide association studies revealed a relationship between SLC16A9 and serum uric acid concentrations (Phipps-Green et al. Citation2016). Genotyping of rs12356193 within SLC16A9 was associated with dl-carnitine and propionyl-l-carnitine concentrations, which in turn were associated with serum UA levels. It implies that MCT9 could indirectly affect extra-renal urate excretion (Kolz et al. Citation2009). Another missense variant of MCT9, rs2242206, significantly increased the risk of renal overload gout but not of renal underexcretion gout. Although the function of MCT9 remains not fully unclear, MCT9 might have a possible physiological role in urate excretion from human intestinal epithelial cells where MCT9 expression is observed (Nakayama et al. Citation2013).

Na+/phosphate co-transporters

The Na+/phosphate co-transporters (NPTs), an SLC17 family of proteins initially characterized as phosphate carriers, mediate the transport of organic anions. Three SLC17 family members associated with serum uric acid concentrations have been identified through genomic analysis, and designated NPT1 (SLC17A1), NPT4 (SLC17A3) and NPT homolog (SLC17A4). Human sodium phosphate transporter 4 (hNPT4) is localized at the apical side of renal tubules and functions as a voltage-driven uric acid transporter. It is likely to act as a common secretion route for both drugs and may play an important role in diuretics-induced hyperuricaemia (Jutabha et al. Citation2010). Actually, NPT4 is present in the small intestine too (Reimer & Edwards Citation2004).

SLC17A4 protein was isolated from a human intestine mucosal. The amino acids sequence shows 48% overall similarity with the renal NPT1 protein (Shibui et al. Citation1999). Since NPT1 is responsible for renal uric acid extrusion, heterologous expression reveals functional characteristics similar to those of NPT1 and NPT4, indicating the possible involvement of the NPT homolog (NPT5) in uric acid extrusion from the intestinal tract (Togawa et al. Citation2012).

Organic anion transporter 10 (OAT10)

The OAT subfamily (SLC22A) constitutes roughly half of the SLC22 transporter family. Most of OATs are highly expressed in the kidneys. Nine OATs were functionally identified (OAT1-7, Oatv1 and URAT1). Some OATs have a strong selectivity for uric acid (Nigam et al. Citation2015). Human organic anion transporter (hOAT10) is highly expressed in the kidney and to a weaker extent in the intestine. From a cyclosporine A-induced hyperuricaemia, hOAT10 was identified as a new uric acid transporter (Bahn et al. Citation2008).

Bacterial uric acid transporters

The intestine is the main site of uricolysis (the degradation of uric acid). Some disappearance of uric acid was observed in cultures of Escherichia coli, Aerobacter aerogenes and Paracolobactrum species, isolated from human feces and from intestinal contents of rats (Lau & Wiseman Citation1964). In the lower intestinal tract, uric acid is exposed to a large population of bacteria that can use the uric acid as a metabolic substrate (Braun & Campbell Citation1989). When uric acid was administered to human subjects by intravenous injections, about 80% of the uric acid was excreted in the urine. If administered by mouth, uric acid was extensively degraded in the gastrointestinal tract, presumably by bacterial action (Wyngaarden & Stetten Citation1953). The ubiquitous nucleobase–ascorbate transporter (NAT/NCS2) family includes more than 2000 members, but only 15 have been characterized experimentally. Among Escherichia coli members, the xanthine permeases XanQ and XanP are functionally known. Of the remaining members, YgfU is closely related in sequence and genomic locus with XanQ. YgfU is a proton-gradient dependent, low-affinity and high-capacity transporter for uric acid (Papakostas & Frillingos Citation2012).

Perspectives

The increasing knowledge of uric acid transporters sheds light on the causes of hyperuricemia. Identification of the renal transporter URAT1 in 2002 paved the way for successive clarification of the uric acid transport system. Novel technologies and various useful public databases have improved our understanding of the physiological roles of transporters. Although the intestinal contribution in uric acid excretion has been recognized over the past 50 years, molecular information of intestinal uric acid transport is obtained quite recently, with ABCG2 being the best characterized to date. The battle to ferret out uric acid transporters hiding in the intestine has just begun.

Actually, the history of transporter discovery in the intestine (from P-gp) is longer than that in the kidney. As outlined in this review, some transporters participating in uric acid transport in the kidney are also expressed in the intestine (). However, are the similar mechanisms of these transporters for uric acid transport in the intestine? There is much more we need to address. Future research will continue to investigate the precise role of the known transporters and to search for candidates related to intestinal uric acid handling. The pathophysiological roles of intestinal uric acid transporters need to be unveiled, especially during renal insufficiency processes. In view of a wider space and a wide variety of bacteria in the intestine, it seems likely that transporters in the intestine would be much more complex than in the kidney. Studying transporter interaction profiles can be critical to fully understand the precise mechanism governing the bi-directional transport of uric acid. The interaction of transporter with other associated proteins has not been functionally confirmed yet, as uric acid transporters represents an important challenge. The entire picture of uric acid metabolism will advance our knowledge and give a wider selection of effective treatments for hyperuricemia.

Table 1. Uric acid transporters in the intestine.

Disclosure statement

The authors report that they have no conflict of interest.

Funding

This project was granted financial support from China Postdoctoral Science Foundation, No. 2015M571253.

References

  • Augustin R, Carayannopoulos MO, Dowd LO, Phay JE, Moley JF, Moley KH. 2004. Identification and characterization of human glucose transporter-like protein-9 (GLUT9): alternative splicing alters trafficking. J Biol Chem. 279:16229–16236.
  • Bahn A, Hagos Y, Reuter S, Balen D, Brzica H, Krick W, Burckhardt BC, Sabolic I, Burckhardt G. 2008. Identification of a new urate and high affinity nicotinate transporter, hOAT10 (SLC22A13). J Biol Chem. 283:16332–16341.
  • Bataille AM, Goldmeyer J, Renfro JL. 2008. Avian renal proximal tubule epithelium urate secretion is mediated by Mrp4. Am J Physiol Regul Integr Comp Physiol. 295:R2024–33.
  • Braun EJ, Campbell CE. 1989. Uric acid decomposition in the lower gastrointestinal tract. J Exp Zool Suppl. 3:70–74.
  • Buzard J, Bishop C, Talbott JH. 1952. Recovery in humans of intravenously injected isotopic uric acid. J Biol Chem. 196:179–184.
  • Campion EW, Glynn RJ, De Labry LO. 1987. Asymptomatic hyperuricemia. Risks and consequences in the Normative Aging Study. Am J Med. 82:421–426.
  • De Bosch BJ, Kluth O, Fujiwara H, Schürmann A, Moley K. 2014. Early-onset metabolic syndrome in mice lacking the intestinal uric acid transporter SLC2A9. Nat Commun. 5:4642.
  • Dehghan A, Köttgen A, Yang Q, Hwang SJ, Kao WL, Rivadeneira F, Boerwinkle E, Levy D, Hofman A, Astor BC, et al. 2008. Association of three genetic loci with uric acid concentration and risk of gout: a genome-wide association study. Lancet. 372:1953–1961.
  • de Oliveira EP, Burini RC. 2012. High plasma uric acid concentration: causes and consequences. Diabetol Metab Syndr. 4:12–18.
  • Doyle LA, Yang W, Abruzzo LV, Krogmann T, Gao Y, Rishi AK, Ross DD. 1998. A multidrug resistance transporter from human MCF-7 breast cancer cells. Proc Natl Acad Sci USA. 95:15665–15670.
  • Dudas PL, Pelis RM, Braun EJ, Renfro JL. 2005. Transepithelial urate transport by avian renal proximal tubule epithelium in primary culture. J Exp Biol. 208:4305–4315.
  • Enomoto A, Kimura H, Chairoungdua A, Shigeta Y, Jutabha P, Cha SH, Hosoyamada M, Takeda M, Sekine T, Igarashi T, et al. 2002. Molecular identification of a renal urate anion exchanger that regulates blood urate levels. Nature. 417:447–452.
  • Estudante M, Morais JG, Soveral G, Benet LZ. 2013. Intestinal drug transporters: an overview. Adv Drug Deliv Rev. 65:1340–1356.
  • Grandvuinet AS, Vestergaard HT, Rapin N, Steffansen B. 2012. Intestinal transporters for endogenic and pharmaceutical organic anions: the challenges of deriving in-vitro kinetic parameters for the prediction of clinically relevant drug-drug interactions. J Pharm Pharmacol. 64:1523–1548.
  • Halestrap AP, Meredith D. 2004. The SLC16 gene family-from monocarboxylate transporters (MCTs) to aromatic amino acid transporters and beyond. Pflugers Arch. 447:619–628.
  • Halestrap AP, Price NT. 1999. The proton-linked monocarboxylate transporter (MCT) family: structure, function and regulation. Biochem J. 343:281–299.
  • Hediger MA, Johnson RJ, Miyazaki H, Endou H. 2005. Molecular physiology of urate transport. Physiology (Bethesda). 20:125–133.
  • Hilgendorf C, Ahlin G, Seithel A, Artursson P, Ungell AL, Karlsson J. 2007. Expression of thirty-six drug transporter genes in human intestine, liver, kidney, and organotypic cell lines. Drug Metab Dispos. 35:1333–1340.
  • Hosomi A, Nakanishi T, Fujita T, Artursson P, Ungell AL, Karlsson J. 2012. Extra-renal elimination of uric acid via intestinal efflux transporter BCRP/ABCG2. PLoS One. 7:e30456.
  • Ichida K, Matsuo H, Takada T, Nakayama A, Murakami K, Shimizu T, Yamanashi Y, Kasuga H, Nakashima H, Nakamura T, et al. 2012. Decreased extra-renal urate excretion is a common cause of hyperuricemia. Nat Commun. 3:764.
  • Iwama M, Kondo Y, Shimokado K, Maruyama N, Ishigami A. 2012. Uric acid levels in tissues and plasma of mice during aging. Biol Pharm Bull. 35:1367–1370.
  • Jutabha P, Anzai N, Kitamura K, Maruyama N, Ishigami A. 2010. Human sodium phosphate transporter 4 (hNPT4/SLC17A3) as a common renal secretory pathway for drugs and urate. J Biol Chem. 285:35123–35132.
  • Krishnan E, Bennett M, Chen L. 2014. Aging, not menopause, is associated with higher prevalence of hyperuricemia among older women. Menopause. 21:1211–1216.
  • Kolz M, Johnson T, Sanna S, Teumer A, Vitart V, Perola M, Mangino M, Albrecht E, Wallace C, Farrall M, et al. 2009. Meta-analysis of 28,141 individuals identifies common variants within five new loci that influence uric acid concentrations. PloS Genet. 5:e1000504.
  • Lau HS, Wiseman RF. 1964. Effect of uric acid on riboflavine production by intestinal coliform bacteria. J Bacteriol. 87:337–340.
  • Lima WG, Martins-Santos ME, Chaves VE. 2015. Uric acid as a modulator of glucose and lipid metabolism. Biochimie. 116:17–23.
  • Mandal AK, Mount DB. 2015. The molecular physiology of uric acid homeostasis. Annu Rev Physiol. 77:323–345.
  • Matsuo H, Ichida K, Takada T, Nakayama A, Nakashima H, Nakamura T, Kawamura Y, Takada Y, Yamamoto K, Inoue H, et al. 2013. Common dysfunctional variants in ABCG2 are a major cause of early-onset gout. Sci Rep. 3:2014.
  • Matsuo H, Takada T, Nakayama A, Chiba T, Shimizu S, Kawamura Y, Nakashima H, Nakamura T, Takada Y, Oikawa Y, et al. 2014. ABCG2 dysfunction causes hyperuricemia due to both renal urate underexcretion and renal urate overload. Sci Rep. 4:3755.
  • Moon C, Zhang W, Ren A, Arora K, Sinha C, Yarlagadda S, Woodrooffe K, Schuetz JD, Valasani KR, de Jonge HR, et al. 2015. Compartmentalized accumulation of cAMP near complexes of multidrug resistance protein 4 (MRP4) and cystic fibrosis transmembrane conductance regulator (CFTR) contributes to drug-induced diarrhea. J Biol Chem. 290:11246–11257.
  • Mueckler M, Thorens B. 2013. The SLC2 (GLUT) family of membrane transporters. Mol Aspects Med. 34:121–138.
  • Nakayama A, Matsuo H, Shimizu T, Ogata H, Takada Y, Nakashima H, Nakamura T, Shimizu S, Chiba T, Sakiyama M, et al. 2013. A common missense variant of monocarboxylate transporter 9 (MCT9/SLC16A9) gene is associated with renal overload gout, but not with all gout susceptibility. Hum Cell. 26:133–136.
  • Nakayama A, Matsuo H, Takada T, Ichida K, Nakamura T, Ikebuchi Y, Ito K, Hosoya T, Kanai Y, Suzuki H, et al. 2011. ABCG2 is a high-capacity urate transporter and its genetic impairment increases serum uric acid levels in humans. Nucleosides Nucleotides Nucleic Acids. 30:1091–1097.
  • Nigam SK, Bush KT, Martovetsky G, Ahn SY, Liu HC, Richard E, Bhatnagar V, Wu W. 2015. The organic anion transporter (OAT) family: a systems biology perspective. Physiol Rev. 95:83–123.
  • Oh JH, Dossetor JB, Beck IT. 1967. Kinetics of uric acid transport and its production in rat small intestine. Can J Physiol Pharmacol. 45:121–127.
  • Oka Y, Tashiro H, Sirasaki R, Yamamoto T, Akiyama N, Kawasugi K, Shirafuji N, Fujimori S. 2014. Hyperuricemia in hematologic malignancies is caused by an insufficient urinary excretion. Nucleosides Nucleotides Nucleic Acids. 33:434–438.
  • Papakostas K, Frillingos S. 2012. Substrate selectivity of YgfU, a uric acid transporter from Escherichia coli. J Biol Chem. 287:15684–15695.
  • Perez-Ruiz F, Dalbeth N, Bardin T. 2015. A review of uric acid, crystal deposition disease, and gout. Adv Ther. 32:31–41.
  • Phay JE, Hussain HB, Moley JF. 2000. Cloning and expression analysis of a novel member of the facilitative glucose transporter family, SLC2A9 (GLUT9). Genomics. 66:217–220.
  • Phipps-Green AJ, Merriman ME, Topless R, Phipps-Green AJ, Merriman ME, Topless R, Altaf S, Montgomery GW, Franklin C, Jones GT, et al. 2016. Twenty-eight loci that influence serum urate levels: analysis of association with gout. Ann Rheum Dis. 75:124–130.
  • Preitner F, Bonny O, Laverrière A, Rotman S, Firsov D, Da Costa A, Metref S, Thorens B. 2009. 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.
  • Reimer RJ, Edwards RH. 2004. Organic anion transport is the primary function of the SLC17/type I phosphate transporter family. Pflugers Arch. 447:629–635.
  • Richette P, Bardin T. 2010. Gout. Lancet. 375:318–328.
  • Roninson IB, Chin JE, Choi KG, Gros P, Housman DE, Fojo A, Shen DW, Gottesman MM, Pastan I. 1986. Isolation of human mdr DNA sequences amplified in multidrug-resistant KB carcinoma cells. Proc Natl Acad Sci USA. 83:4538–4542.
  • Sakurai H. 2013. Urate transporters in the genomic era. Curr Opin Nephrol Hypertens. 22:545–550.
  • Sarkadi B, Ozvegy-Laczka C, Német K, Váradi A. 2004. ABCG2-a transporter for all seasons. FEBS Lett. 567:116–120.
  • Sedykh A, Fourches D, Duan J, Hucke O, Garneau M, Zhu H, Bonneau P, Tropsha A. 2013. Human intestinal transporter database: QSAR modeling and virtual profiling of drug uptake, efflux and interactions. Pharm Res. 30:996–1007.
  • Shibui A, Tsunoda T, Seki N, Suzuki Y, Sugane K, Sugano S. 1999. Isolation and chromosomal mapping of a novel human gene showing homology to Na+/PO4 cotransporter. J Hum Genet. 44:190–192.
  • Smeets PH, van Aubel RA, Wouterse AC, van den Heuvel JJ, Russel FG. 2004. Contribution of multidrug resistance protein 2 (MRP2/ABCC2) to the renal excretion of p-aminohippurate (PAH) and identification of MRP4 (ABCC4) as a novel PAH transporter. J Am Soc Nephrol. 15:2828–2835.
  • So A, Thorens B. 2010. Uric acid transport and disease. J Clin Invest. 120:1791–1799.
  • Sorensen LB. 1965. Role of the intestinal tract in the elimination of uric acid. Arthritis Rheum. 8:694–706.
  • Sorensen LB, Levinson DJ. 1975. Origin and extrarenal elimination of uric acid in man. Nephron 14:7–20.
  • Susic D, Frohlich ED. 2015. Hyperuricemia: a biomarker of renal hemodynamic impairment. Cardiorenal Med. 5:175–182.
  • Takada T, Ichida K, Matsuo H, Nakayama A, Murakami K, Yamanashi Y, Kasuga H, Shinomiya N, Suzuki H. 2014. ABCG2 dysfunction increases serum uric acid by decreased intestinal urate excretion. Nucleosides Nucleotides Nucleic Acids. 33:275–281.
  • Togawa N, Miyaji T, Izawa S, Omote H, Moriyama Y. 2012. A Na+-phosphate cotransporter homologue (SLC17A4 protein) is an intestinal organic anion exporter. Am J Physiol Cell Physiol. 302:C1652–C1660.
  • Van Aubel RA, Smeets PH, Van Den Heuvel JJ, Russel FG. 2004. 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 Renal Physiol. 288:F327–F333.
  • Vaziri ND, Freel RW, Hatch M. 1995. Effect of chronic experimental renal insufficiency on urate metabolism. J Am Soc Nephrol. 6:1313–1317.
  • Vitart V, Rudan I, Hayward C, Gray NK, Floyd J, Palmer CN, Knott SA, Kolcic I, Polasek O Graessler J, et al. 2008. SLC2A9 is a newly identified urate transporter influencing serum urate concentration, urate excretion and gout. Nat Genet. 40:437–442.
  • Woodward OM, Köttgen A, Coresh J, Boerwinkle E, Guggino WB, Köttgen M. 2009. Identification of a urate transporter, ABCG2, with a common functional polymorphism causing gout. Proc Natl Acad Sci USA. 106:10338–10342.
  • Wyngaarden JB, Stetten D Jr. 1953. Uricolysis in normal man. J Biol Chem. 203:9–21.
  • Yano H, Tamura Y, Kobayashi K, Tanemoto M, Uchida S. 2014. Uric acid transporter ABCG2 is increased in the intestine of the 5/6 nephrectomy rat model of chronic kidney disease. Clin Exp Nephrol. 18:50–55.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.