Open access peer-reviewed chapter - ONLINE FIRST

Role of Transporters in Hyperuricemia

Written By

Siqi Zhang, Jingwen Wang, Shuangxiang Wang, Zekai Dai, Lei Zhang and Feng Xue

Reviewed: 25 April 2024 Published: 22 May 2024

DOI: 10.5772/intechopen.114991

Cell Communication and Signaling in Health and Disease IntechOpen
Cell Communication and Signaling in Health and Disease Edited by Thomas Heinbockel

From the Edited Volume

Cell Communication and Signaling in Health and Disease [Working Title]

Dr. Thomas Heinbockel

Chapter metrics overview

45 Chapter Downloads

View Full Metrics

Abstract

Hyperuricemia (HUA) is a metabolic disorder characterized by elevated serum uric acid levels exceeding the body’s metabolic limit. In the past two decades, the prevalence of this disease has shown an increasing trend and is becoming more common in younger individuals. As a metabolic disease, hyperuricemia has been found to correlate with cardiovascular diseases, renal diseases, and metabolic syndrome. Various complex metabolic processes are involved in the pathological process in the elevation of uric acid. Transporters are one of the most important families controlling the metabolism of uric acid. The vast majority of cases of hyperuricemia are caused by insufficient uric acid excretion and excessive reabsorption by the kidneys. Therefore, limiting the reabsorption of transport proteins is key to lowering uric acid levels. This chapter will revisit the basic situation of hyperuricemia and summarize the known mechanisms of transport proteins in HUA, as well as the therapeutic approaches developed for these transport proteins.

Keywords

  • hyperuricemia
  • uric acid metabolism
  • transport proteins
  • molecular mechanism
  • therapeutic strategies

1. Introduction

1.1 Hyperuricemia

1.1.1 Concept

Hyperuricemia, characterized by elevated uric acid (UA) levels in the bloodstream, can arise from both exogenous (dietary) and endogenous purine conversion processes, primarily orchestrated by the liver and intestines [1]. Although kidneys and intestines typically excrete uric acid, an increase in production or impaired elimination pathways may lead to its accumulation, resulting in hyperuricemia. Elevated uric acid concentrations can foster urate crystal formation, which deposits in joints and tissues, triggering diseases like gout.

According to the “2021 Guidelines for the Diagnosis and Treatment of Hyperuricemia and Gout,” regardless of gender and under normal purine diet conditions, a fasting blood uric acid concentration exceeding 420 μmol/L on two separate occasions indicates hyperuricemia [2]. Epidemiological studies demonstrate that in males, hyperuricemia is identified by serum uric acid levels exceeding 7.0 mg/dL (>420 μmol/L), while in females, it is noted at levels surpassing 6.0 mg/dL (>360 μmol/L) [3]. These parameters offer valuable insights into the clinical assessment of hyperuricemia, forming a basis for diagnosis and management.

1.1.2 Epidemiology of hyperuricemia

During the years 2015–2016, the prevalence of hyperuricemia among adult men and women in the United States stood at 20.2 and 20.0%, respectively, and has demonstrated stability over the preceding decade [4]. As of 2021, research has unveiled a range of hyperuricemia prevalence rates across regions, with the United States displaying figures spanning from 11.3 to 47%, Europe exhibiting rates between 11.9 and 25.0%, Japan registering a prevalence of 26.8%, and China showing rates fluctuating between 13.1 and 13.3% [5]. Recent epidemiological studies suggest a relatively high prevalence of hyperuricemia with an upward trajectory [6].

1.1.3 Relationship between hyperuricemia and other diseases

Hyperuricemia is increasingly recognized as a significant health concern, often dubbed the “fourth high,” alongside elevated blood sugar, high blood lipids, and hypertension. Extensive research highlights the link between hyperuricemia and various medical conditions, such as gout, chronic kidney disease, cardiovascular diseases, and diabetes. The presence of hyperuricemia, characterized by the deposition of monosodium urate (MSU) crystals and the activation of inflammatory pathways, poses a primary risk factor for gout attacks [7].

Moreover, hyperuricemia contributes to an increased incidence of coronary heart disease (CHD) and impacts the progression and prognosis of heart failure patients [89]. Investigations reveal a heightened prevalence of diabetes among individuals with hyperuricemia and gout, emphasizing the potential for controlling uric acid levels to partially prevent early-stage diabetes [10].

In the context of chronic kidney disease (CKD), hyperuricemia assumes a significant clinical role in disease onset. Uric acid itself may pose harm to CKD patients by amplifying inflammation and fostering CKD progression. Elevated serum uric acid levels can induce kidney damage through crystal-dependent pathways and non-crystal-dependent mechanisms, including inflammation, oxidative stress, and hemodynamic changes [11]. These findings underscore the multifaceted impact of hyperuricemia on various health conditions, highlighting its relevance in clinical contexts.

Exploring hyperuricemia is crucial for unraveling its associations with gout, uric acid-related kidney diseases, metabolic syndrome, and cardiovascular diseases, among others. A thorough investigation into the causes, mechanisms, and contributing factors of hyperuricemia is essential for enhancing our comprehension and enabling early diagnosis and intervention. This comprehensive research is pivotal for developing more effective treatment strategies, ultimately improving patient outcomes and contributing to overall population health enhancement.

1.2 Mechanisms of uric acid metabolism

Purine sources within the human body are distinguished into endogenous and exogenous categories, with 80% originating from endogenous synthesis or the oxidative breakdown of nucleic acids. The residual 20% is derived from exogenous purines, predominantly influenced by dietary factors, with notable contributions from animal proteins. Uric acid formation, arising from purine catabolism, is chiefly orchestrated through a sequence of enzymatic reactions involving xanthine oxidase, primarily occurring in the liver and intestines [2].

Under normal physiological conditions, the synthesis and excretion of uric acid maintain a dynamic equilibrium, with the kidneys serving as the principal regulatory factor for uric acid excretion [12, 13]. Approximately 70% of UA processing takes place through renal mechanisms, involving initial free filtration at the renal glomerulus, followed by secretion and reabsorption facilitated by renal tubules and associated transport proteins. The majority of circulating urate exists in a free form, exhibiting a protein binding rate of less than 5%, rendering them readily filterable by the renal glomerulus. Nevertheless, as much as 90% of urate may undergo reabsorption, returning to the bloodstream [3]. The remaining 30% of UA traverses the intestines, where transport proteins and microbial metabolism collectively contribute to its elimination (Figure 1) [14, 15, 16].

Figure 1.

Mechanisms of UA production and excretion. Figure legends: Endogenous purines produced by cell metabolism or apoptosis and exogenous purines produced by ingested food constitute an important source of purines in the human body, mainly in the liver after a series of enzymatic reactions with xanthine oxidase as an important rate-limiting enzyme to generate uric acid, about 30% of uric acid through the intestinal transport proteins and the role of the bacterial flora to the body; about 70% of uric acid through the renal filtration, after filtration, about 90% is reabsorbed back into the blood circulation with the assistance of transport proteins, and the remaining 10% of uric acid is excreted. About 70% of the uric acid is filtered through the kidneys, about 90% of the filtered uric acid is reabsorbed back into the blood circulation with the assistance of transport proteins, and the remaining about 10% of the uric acid is excreted from the body.

1.3 Transport proteins

Transporter proteins, a class pivotal in orchestrating the movement of molecules or ions within living organisms, exhibit their functional roles on cell membranes or within the cellular membrane system. Through specific structures and mechanisms, these proteins facilitate the transfer of specific molecules from one location to another. The human genome presently encompasses more than 400 annotated membrane transporter proteins [17]. Following the guidelines established by the HUGO Gene Nomenclature Committee, these transporter proteins are mainly categorized into the solute carrier (SLC) and ATP-binding cassette (ABC) superfamily [18].

The SLC proteins, constituting an exceptionally diverse superfamily with over 400 members, exhibit extensive functional variability [19]. Concurrently, the ABC transporter proteins, totaling 48 members within the transmembrane transporter protein superfamily, actively engage in transporting diverse biological substrates. The multifaceted functionalities of these transporter proteins underscore their widespread involvement in numerous facets of human biology [20].

In this context, we give a brief overview of the principal transporter proteins identified in different organs, including kidneys, liver, and intestines (Table 1).

The transporting familyTransporterDominant locationPrimary functionReferences
Solute carrier transportersOATPLiver, kidney, intestineOrganic anion transporting polypeptide[21, 22]
NTCPLiverSodium taurocholate cotransporting[23]
OATLiver, kidneyOrganic anion transporters[24]
OCTLiver, kidneyOrganic Cation Transporter[25]
MATELiver, kidneyMultidrug and toxic compound extrusion[26]
URAT1Kidney, intestineUrate transporter[27]
OCTNKidney, intestineOrganic Cation Transporters Novel[28]
OSTα/βLiverBile acids, steroid metabolites and drugs can be transported into and out of cells[29]
PEPTIntestineTransporting peptides or proteins between intracellular and extracellular spaces.[30]
PMATIntestineCapable of transporting monoamine neurot ransmitters with low affinity and high capacity[31]
MCTLiver, intestineCatalyze the proton linking and transport of monocarboxylate across plasma membranes[32, 33]
GLUTIntestineGlucose is transported along the concentration gradient in a facilitated diffusion manner[34]
Pgp (MDR)Kidney, intestineMultidrug resistant protein[35]
ATP-Binding Cassette (ABC) TransporterBCRP (ABCG 2)Liver, kidney, intestineThe breast cancer resistance protein[36]
MRPLiver, kidney, intestineCauses multidrug resistance in tumor cells by squeezing anticancer drugs out of cells[11]
BSEPLiverEliminating unconjugated and conjugated bile acids/salts from hepatocytes into bile[37]

Table 1.

Transport proteins in the liver, kidneys, and intestines.

1.3.1 Transport proteins in the kidneys

In the renal domain, numerous SLC transporter proteins are present, including organic anion transporters (OAT 1/2/3/4), organic cation transporters (OCT 2/3), organic anion-transporting polypeptides (OATP 4C 1), multidrug and toxin extrusion proteins (MATE 1/2-K), urate anion exchanger protein (URAT1), and organic cation/carnitine transporters (OCTN 1/2), among others. Specifically, the SLC22 transporter protein family encompasses OAT, OCT, OCTN, and URAT1 [38].

In the renal environment, ABC transporter proteins are also present, including multidrug resistance protein (MDR 1 or PGP), breast cancer resistance protein (BCRP), and multidrug resistance-associated proteins (MRP 2/4). These transporters utilize the energy derived from ATP hydrolysis to facilitate the movement of molecules across cellular membranes [39].

1.3.2 Transport proteins in the liver

In the hepatic milieu, SLC transporter proteins prominently feature organic anion-transporting polypeptide (OATP), sodium-dependent taurocholate cotransporting polypeptide (NTCP) [23], and organic cation transporter proteins (OCT) exemplified by OCT1 and OCT2 [40], along with organic solute transporter alpha/beta (OSTα/β). OATP assumes a pivotal role as a major drug carrier protein, facilitating sodium-independent hepatic uptake of organic anions [41]. Transport facilitated by OSTα/β exhibits bidirectionality (uptake or efflux), operates in an ATP-independent manner, and relies on the electrochemical gradient [42].

The ABC transporter family primarily comprises multidrug resistance proteins (MRP) such as MRP2, multidrug resistance proteins (MDR) including MDR1/3/4, bile salt export pump (BSEP), and breast cancer resistance protein (BCRP/ABCG2). These transporter proteins are strategically positioned on the apical side of hepatocytes, emphasizing their role in hepatic transport processes [28].

1.3.3 Transport proteins in the intestine

Among the intestinal transport proteins, the extensively studied ones predominantly belong to the ABC family, featuring efflux transport proteins such as P-glycoprotein (P-gp), multidrug resistance protein 2 (MRP2), and breast cancer resistance protein (BCRP). Simultaneously, SLC transport proteins in the intestinal realm primarily encompass facilitated diffusion transport proteins (GLUT) [43], peptide transport proteins (PEPT), organic cation transporter proteins (OCT), plasma membrane monoamine transporter (PMAT), organic anion-transporting polypeptide (OATP), and monocarboxylate transporter proteins (MCT) [44]. This diversity in transport proteins underscores the intricacies of intestinal transport mechanisms, implicating their significance in various physiological processes.

1.4 Challenges

Membrane transport proteins not only play pivotal roles in physiology but also wield significant influence in pharmacology, shaping the absorption, distribution, and excretion dynamics of nutrients, signaling molecules, metabolic end products, and pharmaceuticals. However, the study of membrane transport proteins encounters several challenges:

  1. Despite achieving successful structural resolutions for certain transport proteins, many, particularly intricate membrane proteins, remain only partially understood in terms of their structures.

  2. The dynamics and regulatory intricacies of transport proteins manifest complexities within distinct intracellular and extracellular milieus, necessitating a deeper comprehension of these nuanced details.

  3. Interactions between transport proteins and other proteins, ligands, or cargo substances are commonplace, yet the precise details of these interactions warrant further investigative efforts.

  4. Although certain diseases are linked to the aberrant function of transport proteins, the exact roles they play in these diseases remain inadequately understood.

  5. While some drugs target transport proteins, the development of specific and efficient drugs remains a formidable challenge, particularly given the unique structural features of membrane proteins.

Addressing these issues mandates interdisciplinary research approaches that seamlessly blend experimental and computational techniques, fostering a comprehensive understanding of the biological functions and regulatory mechanisms governing transport proteins.

Advertisement

2. Mechanisms and roles of transport proteins in HUA

2.1 Transport proteins in HUA

Urate transporter proteins stand as pivotal targets in the research and drug development for hyperuricemia.

The kidney plays a predominant role in maintaining serum urate levels through its excretory function, eliminating approximately 70% of daily urate production. Urate metabolism in the kidneys involves three processes: filtration, secretion, and reabsorption, primarily occurring in the renal proximal tubules. Apart from filtration, urate transporter proteins participate in the remaining processes [4546]. After filtration, a large portion of urate ions are reabsorbed, constituting an important component of serum uric acid levels in the body. Urate transporter 1 (URAT1) and glucose transporter 9 (GLUT9) are important transport proteins involved in urate reabsorption. In addition to these, there are also organic anion transport proteins OAT4 and OAT10 in the OAT family that play a role in urate reabsorption [47]. Transporter proteins such as ABCG2, OAT1, and OAT3 present on the epithelial cells of the renal proximal tubules serve as important outlets mediating urate excretion, while ABCC4, a known organic anion transporter protein, also plays a role in urate excretion, though its specific location remains unclear.

The kidney was previously believed to be the sole pathway for urate excretion; however, it is estimated that approximately one-third of urate is excreted into the intestine, where it undergoes further metabolism by intestinal bacteria [15]. Intestinal secretion is considered a significant factor in extra-renal urate elimination [48]. Intestinal transporter proteins play crucial roles in urate excretion, yet the molecular characteristics of urate transporter proteins in the intestine have not been extensively studied to date. Members of the ABC family may play a role in intestinal urate excretion, with ABCG2 located on the apical membrane of intestinal epithelial cells serving as a crucial outlet for intestinal urate excretion [48]. Given the high expression of ABCG2 in the intestine, it may primarily facilitate intestinal urate excretion rather than renal excretion (Figure 2) [49].

Figure 2.

UA transport proteins in the kidneys and intestines. Figure legends: (a) uric acid enters the kidney and undergoes filtration in the renal capsule, then excretion and reabsorption via transporter proteins in the epithelial cells of the proximal tubule of the kidney. URAT1, GLUT9, OAT4, and OAT10 are responsible for the reabsorption of UA, and ABCG2, ABCC4, OAT1, and OAT3 are responsible for the excretion of UA; (b) a small portion of uric acid is metabolized in the intestinal epithelial cells and is known to excrete UA by ABCG2, and the rest of the intestinal-associated transport proteins need to be explored further.

2.2 Key transporters

Elevated serum urate concentration stands as the paramount risk factor contributing to the onset of hyperuricemia. The journey of urate is intricately orchestrated within the renal landscape: initially freely filtered at the renal glomerulus, followed by reabsorption in the S1 segment of the proximal tubule, and subsequent secretion, potentially accompanied by reabsorption, in the S2-S3 segments of the proximal tubule [50]. A fractional portion of urate undergoes metabolism in the intestines, adding another layer of complexity to its regulation.

Through genetic approaches, an array of renal and gastrointestinal transport proteins with a significant affinity for urate have been unearthed. Notable among these are the SLC transporter protein genes, namely SLC22A12 and SLC2A9, along with the ABC transporter protein gene ABCG2. Remarkably, variations within just these three genes contribute to a substantial 5% of the variability observed in serum urate levels, surpassing the cumulative impact of all other genetic variations. These findings illuminate the intricate genetic landscape underpinning urate regulation and underscore the pivotal role played by specific transport proteins in shaping serum urate dynamics [51, 52]. Serum uric acid is regulated by urate transport proteins in the kidneys and intestines, with most of the uric acid being reabsorbed back into the bloodstream. Therefore, we will focus on the characteristics of three important transport proteins: URAT1 and GLUT9, which are associated with uric acid reabsorption, and ABCG2, which is involved in uric acid excretion [53].

2.2.1 URAT1

URAT1 stands as a pivotal apical urate exchanger within the proximal renal tubules, wielding a central role in the intricate physiological homeostasis of urate [54]. Comprising 12 transmembrane domains and 555 amino acid residues, URAT1 is encoded by the SLC22A12 gene, sharing a noteworthy 30% homology with the rat organic cation transporter 1 [55]. Originally uncovered during the exploration of organic anion transporter-like (OAT-like) molecules in the gene database, URAT1’s functional aspects were revealed through expression studies in African clawed frog oocytes [56].

URAT1, with its high affinity for uric acid, exhibits specific expression on the luminal side of the proximal renal tubules. Responsible for transporting uric acid from the lumen of the proximal renal tubule to the apical membrane through the transmembrane electric potential gradient, URAT1 plays a crucial role in mediating urate salt uptake through exchange with organic anions (such as lactate and nicotinate) and inorganic anions Cl-, thereby contributing significantly to the regulation of human serum uric acid levels. In one study, functional analysis of rat Urat1 was conducted using isolated brush border membrane vesicles (BBMVs) prepared from rat kidneys [57]. It was observed that urate uptake by Urat1 exhibited Cl − dependency, with an approximately 57% increase in Km value. Additionally, research conducted by expressing URAT1 in African clawed frog oocytes revealed that the application of inorganic anions Br and I also significantly accelerates urate uptake. Furthermore, it was found that URAT1-mediated urate uptake is not pH-dependent (i.e., no affinity for OH−) [56]. Another study, involving the cloning characterization of human SMCT2 (SLC5A12) and its expression in mouse kidney slices unveiled the remarkable ability of both low-affinity (SMCT2) and high-affinity (SMCT1) lactate transporters to efficiently assist in lactate reabsorption in the proximal tubules of the kidney [58]. Immunofluorescence staining vividly highlighted the intense expression of SMCT2 and SMCT1 in different segments of the proximal renal tubules, suggesting that Urat1 activity might be expedited through preloading lactate.

Certain uricosuric agents, including probenecid, sulfinpyrazone, benzbromarone, and losartan, exert their influence by inhibiting urate reabsorption, binding to URAT1, and promoting urate excretion [59]. Noteworthy are the implications of mutations in URAT1, which can precipitate hypouricemia in humans [56]. Japan, in particular, reports a heightened prevalence of hereditary renal hypouricemia, with approximately 90% of Japanese hypouricemia patients attributable to non-functional variants of URAT1 [60]. Mouse studies further corroborate URAT1’s significance, revealing that knocking out the URAT1 gene induces mild hypouricemia and heightened urate salt excretion [61]. Another study indicated that Urat-Uox double knockout mice exhibit elevated ATP degradation, with uric acid excretion levels increasing 25 times that of humans, leading to decreased serum uric acid levels [62]. These findings underscore URAT1’s indispensable role in urate regulation and its potential as a therapeutic target for modulating serum urate levels.

2.2.2 GLUT9

GLUT9, encoded by the SLC2A9 gene, stands as a distinguished member within the glucose transporter (GLUT) family. Its structure aligns with the type II glutamine subtype, featuring an intricate arrangement of 12 transmembrane domains. [63]. Initially identified for its role as a glucose and/or fructose transporter, rather than a urate transporter [64], GLUT9 shares the characteristic inhibition by cell relaxin B observed in other GLUT family members. Recent revelations, however, illuminate GLUT9’s role as a high-capacity urate transporter, pivotal for the basolateral transport of urate in the proximal tubule. This function contributes to urate reabsorption by facilitating the transport of glucose [65, 66]. The findings revealed that the urate transport rate of SLC2A9 is 45 to 60 times faster than that of glucose, and it was demonstrated that the urate transport mediated by SLC2A9 is primarily facilitated by glucose, with fructose playing a lesser role [66].

GLUT9 manifests in two distinct isoforms, SLC2A9-L (540 amino acids) and SLC2A9-S (512 amino acids). Research by Kimura et al. unveils the unique locations of these isoforms, with SLC2A9-L situated on the basolateral membrane of the proximal tubule and SLC2A9-S on the apical membrane of the collecting duct [67]. Intriguingly, certain kinetic properties of the GLUT9 isoforms defy differentiation. Additionally, GLUT9, owing to its ability to induce depolarization, functions more akin to an electronic transporter rather than a urate-anion exchanger [67, 68, 69]. Preitner et al.’s study revealed that knockout of the SLC2A9 gene in mice resulted in elevated blood creatinine levels and a 20 to 30-fold increase in urate salt excretion rates. In male G9KO mice, the excretion fraction approached approximately 100%, while in female G9KO mice, the urate salt excretion fraction reached approximately 150%. These findings suggest inhibited urate salt reabsorption and increased tubular excretion in both male and female G9KO mice [70].

2.2.3 ABCG2

ABCG2, also known as the human breast cancer resistance protein (BCRP), stands among the trio of human ATP-binding cassette (ABC) transporter proteins, ubiquitously distributed throughout the body. Its role extends to facilitating the cellular efflux of a diverse array of chemically and structurally varied compounds [12, 71]. Revered as a “gatekeeper” in virtue of its function and localization, ABCG2 stands guard, thwarting the passage of endogenous or exogenous toxins and foreign substances across biological barricades into delicate tissues [49].

Expressed in various tissues, ABCG2 graces the luminal membranes of renal tubules and the intestines, where it orchestrates the secretion of an array of compounds, including urate. Notably, ABCG2 assumes a prominent role in urate excretion within the intestines, surpassing its involvement in other tissues, thus emerging as the primary conduit for extra-renal urate elimination [12, 52].

Research has illuminated the expansion of extra-renal urate salt excretion as renal function declines, highlighting its pivotal role in preserving urate homeostasis, especially in individuals grappling with renal insufficiency [72]. Notably, in a rat model of 5/6 nephrectomy, despite diminished uric acid excretion, serum uric acid levels remained unchanged, concurrently with a notable rise in intestinal ABCG2 levels [73]. These findings underscore ABCG2’s pivotal role in intestinal urate excretion, implying that compromised intestinal urate excretion due to ABCG2 dysfunction may underlie one of the mechanisms of hyperuricemia.

Moreover, mutations or dysfunction of ABCG2 may precipitate hyperuricemia or gout [74, 75]. ABCG2 knockout mice exhibit diminished intestinal urate excretion and heightened plasma urate concentrations [12]. A recent study has confirmed that the common ABCG2 variant Q141K, identified in human intervention cohort studies, is associated with an increased risk of hyperuricemia and gout. Additionally, the research team observed significant hyperuricemia and metabolic changes in male mice carrying the direct homologous Q140K Abcg2 variant. These mice exhibited elevated urinary sodium excretion fractions (FeNa+) and decreased glomerular filtration rates (GFR). Notably, there was a substantial reduction in ABCG2 abundance (from 78 to 44%) and severe functional defects observed in the intestines of these mice [52].

2.3 Transporters in the HUA network

The exchange of uric acid is mediated by various molecules expressed in the epithelial cells of the renal proximal tubules and intestinal epithelial cells, primarily including GLUT9, URAT1, and human ABCG2, OAT1, OAT3, and OAT4 (Figure 3).

Figure 3.

UA transport in the network of hyperuricemia. Figure legends: UA exchange occurs predominantly in renal and intestinal epithelial cells, where complex substance exchanges mediated by UA excretory and reabsorptive transport proteins occur to form a hyperuricemia network.

URAT1 facilitates the reabsorption of uric acid in the proximal tubular lumen by exchanging monovalent carboxylate anions or organic anions (such as lactate, pyrazinecarboxylate PZA, and nicotinate) in the epithelial cells [24, 47, 56, 76]. The intracellular concentration of these anions mainly depends on Na + −dependent uptake from the same glomerular ultrafiltration [77]. GLUT9 is a high-capacity electrogenic transporter that significantly activates membrane depolarization (uptake of extracellular [K + -Cl−] in the medium), with a specific ability to transport uric acid unidirectionally through a voltage-dependent manner, seemingly influenced by extracellular Cl-concentration but independent of Na + transmembrane gradients [78]. OAT4 appears to facilitate the reabsorption of substrates in urine through exchange with dicarboxylates (such as succinate) or hydroxyl ions [13, 77, 79]. OAT10 mediates the exchange of urate-PZA and urate-nicotinate. ABCG2 and ABCC4 utilize ATP hydrolysis to expel uric acid from the cell, and the urate efflux mediated by both depends on the intracellular urate concentration [80, 81]. While genetic variations in human MRP4 have not been linked to impacting serum uric acid levels, genetic variations in human ABCG2 have emerged as major factors in human hyperuricemia. OAT1 and OAT3 mediate the movement of substrate molecules from the blood into cells through dicarboxylate/organic anion exchange (Figure 3) [61, 82].

In addition to the previously mentioned transporter proteins, emerging research has identified associations between uric acid excretion and certain related proteins. A comprehensive investigation re-evaluated the tissue distribution of the orphan transporter protein hORCTL3 through RT-PCR, revealing robust expression in the kidneys. This confirmed hORCTL3 as a low-affinity UA transporter [83]. Notably, mutant mice with diminished expression of renal sodium/lactate cotransporter proteins (SLC5A8 and SLC5A12) were found to display impaired uric acid reabsorption, resulting in an approximately eightfold elevation in UA levels [84]. Intriguingly, the expression of URAT1, a transporter protein responsible for the apical uptake of uric acid in the renal proximal tubules, remained unaltered. These findings provide compelling in vivo evidence for a functional link between lactate reabsorption and uric acid reabsorption in the kidneys. The murine ortholog of URAT1, the renal-specific transporter protein RST, is predominantly situated on the apical surface of proximal tubules in the kidneys [55, 56, 85, 86, 87]. While exhibiting significantly reduced affinity when expressed in oocytes, RST does demonstrate uric acid transport capabilities. Knockout C57BL/6Jmice lacking RST exhibits a substantial increase in urinary uric acid (approximately two folds), underscoring the involvement of RST in renal uric acid reabsorption. However, despite the absence of RST, a notable fraction of reabsorption persists, hinting at the presence of unknown transporter proteins that contribute significantly to uric acid reabsorption [61].

Advertisement

3. HUA treatment options targeting transporters

3.1 Drug research targeting HUA-related transport proteins

When urate is excreted from the kidneys, approximately 90% of urate filtered by the renal glomeruli is reabsorbed back into the bloodstream, with only 10% being excreted by the kidneys. Therefore, inhibiting urate reabsorption-related transport proteins is key to uric acid-lowering therapy [54]. In individuals with hyperuricemia, the predominant cause often stems from inadequate urate excretion [14]. Agents designed to enhance urate excretion primarily operate by inhibiting the reabsorption of urate in the proximal renal tubules. Notable transport proteins implicated in urate reabsorption encompass URAT1 and GLUT9. Understanding and modulating the activity of these proteins offer potential avenues for therapeutic interventions aimed at promoting efficient urate elimination (Table 2).

MedicationAnimal models target proteinsPharmacokineticsAdverse drug reactionsMajor contraindications
ProbenecidURAT1no result foundhypersensitivity reactions, rash, gastrointestinal discomfort, gastrointestinal irritation, skin reactions, anorexia.advanced kidney failure, uric acid kidney stones, blood urea, urolithiasis
BenzbromaronePatients URAT1In subjects with CYP2C9*1/1 and CYP2C91/3 genotypes, the plasma concentration of benzbromarone reached its maximum 2 hours after administration, while in subjects with the CYP2C93/3 genotype, it reached the maximum plasma concentration 6 hours after administrationHepatotoxicity, allergy, gastrointestinal discomfort, abnormal liver function, urolithiasis.Individuals with impaired liver and kidney function, those allergic to benzbromarone, patients with uric acid kidney stones, pregnant or potentially pregnant women
LesinuradVolunteers URAT1Mild, moderate, and severe renal impairment increased the plasma concentration of levosulpiride by 34, 54–65, and 102%, respectivelyNephrotoxicity, headache, flulike symptoms, elevated creatinine levels, gastroesophageal refluxSevere renal failure, tumor lysis syndrome, Lesch- Nyhan syndrome
Dotinuradpatients URAT1Regardless of dose, age, and gender, the half-life (T1/2) is approximately 10 hours, with only mild to moderate renal impairment and hepatic impairment observedElevated serum creatinine levels, renal injury, and the potential to lead to the formation of stonesno result found
SulpirideURAT1no result foundNausea, vomiting, abdominal pain, diarrhea, anemia, rashno result found

Table 2.

Limitations of marketed drugs targeting UA reabsorption-related transport proteins.

3.1.1 URAT1

Approved clinical URAT1 inhibitors for urate reabsorption, including probenecid, benzbromarone, sulfinpyrazone, and lesinurad, represent valuable tools in managing hyperuricemia [88, 89]. However, their usage is not without challenges, as these drugs may exhibit varying degrees of side effects and toxicity [90]. Probenecid, while effective in weakening active urate reabsorption, may lead to undesirable effects such as rash, gastrointestinal irritation, hypersensitivity reactions, and hemolytic anemia [91, 92]. Benzbromarone, an efficient URAT1 inhibitor, offers a reduction in serum uric acid levels in CKD patients but demands caution in those with an estimated glomerular filtration rate (eGFR) <30 mL/min due to potential liver toxicity, along with risks of uric acid kidney stones and bone marrow suppression [69, 88, 8991]. Sulfinpyrazone, another option, is associated with adverse effects like nausea, vomiting, abdominal pain, diarrhea, anemia, and rash [93]. Lesinurad, although effective, presents dose-dependent renal toxicity and potential cardiovascular risks, as evidenced by clinical trials [94, 95].

The pursuit of novel urate excretion agents has led to innovative approaches. Utsumi Junichiro and colleagues, for instance, devised Dotinurad (FYU-981), a phenol derivative with enhanced urate-lowering activity [96]. Dotinurad, approved in Japan, addresses structural limitations observed in benzbromarone, proving more effective in reducing plasma UA levels and exhibiting significant inhibitory activity against URAT1-mediated urate transport. Additionally, Philip K. Tan and colleagues explored the potential of verinurad, a novel, potent URAT1 inhibitor [97]. Verinurad’s specificity, demonstrated through competitive binding assays, offers promise for developing highly efficient and specific drugs to treat hyperuricemia.

Natural products, including phenolic compounds, terpenes, and fatty acids, have emerged as intriguing avenues for reducing URAT1 activity. Their potential to lower urate levels has garnered attention, providing a rich field for further exploration in the quest for effective hyperuricemia management [98, 99, 100].

3.1.2 GLUT9

The focus on URAT1 as the primary driver of urate reabsorption has led to limited exploration of drug development targeting the GLUT9 transporter for hyperuricemia treatment, with most studies conducted in conjunction with URAT1 [97]. However, recent research has identified promising compounds with potential anti-hyperuricemic effects and nephroprotective small molecule drugs and traditional Chinese medicine extracts.

Small molecule drugs include oxyberberine (OBR) and betulinic acid. In a study led by Zhong Linjiang and colleagues, OBR demonstrated effectiveness in countering hyperuricemia and protecting the kidneys in hyperuricemic mice [101]. OBR, administered orally, downregulated the renal expression of URAT1, GLUT9, NOD-like receptor 3 (NLRP3), and other factors at both mRNA and protein levels, showcasing its potential as a therapeutic agent. Betulinic acid, a flavonoid compound found in honey, propolis, and mushrooms, exhibited anti-hyperuricemic effects in a rat model induced by high-fructose syrup feeding [102]. Research by Zhang and colleagues explored the mechanism of action of salicin in combating hyperuricemia. The results indicate that salicin exhibits anti-hyperuricemic effects in a rat model of hyperuricemia induced by high-fructose syrup feeding. It significantly downregulates the protein expression of URAT1 and GLUT9 while upregulating the protein expression of OAT1 and human ATP-binding ABCG2.

Traditional Chinese medicine drugs include palmatine (Pal) and apigenin (API). Pal is a major alkaloid from Coptis chinensis, traditionally used for hyperuricemia-related diseases, and showed promise in a hyperuricemia mouse model [103]. Pal effectively reduced the protein levels of GLUT9 and URAT1 while the group Pal (100 mg/kg) increased the expression levels of OAT1 and ABCG2 by nearly twofold compared to the HUA group, suggesting its potential therapeutic role in hyperuricemia. API is a natural flavonoid, demonstrated significant anti-hyperuricemic effects in mice with hyperuricemia nephropathy (HN) [104]. API promoted uric acid excretion, inhibited URAT1 and GLUT9, and showed competitive inhibition in vitro, highlighting its potential as a treatment for hyperuricemia.

Compounds like BDEO (Compound 9), dual inhibitor 83 (Compound 21), and CDER167 (Compound 27) exhibited dual inhibitory effects on URAT1 and XO or GLUT9 [105, 106, 107]. These compounds demonstrated excellent uric acid-lowering capabilities with higher safety profiles, suggesting potential therapeutic avenues for hyperuricemia.

3.1.3 Summary

Presently, there is a growing inclination within the industry toward the development of pharmaceuticals featuring dual-action mechanisms or employing combination therapy approaches. Notable examples in this domain include ACQT-1127 and THDBH151, both functioning as dual inhibitors targeting xanthine oxidase and URAT1 [108]. A pioneering initiative by Westlake Therapeutics involves the utilization of its REDx platform to craft a cell-based therapy [109, 110]. Engineered red blood cells serve as carriers for this innovative approach, specifically designed to target both uricase (UOX) and URAT1. Researchers across the field share a common commitment to ensuring the safety and effectiveness of these developments, particularly in the pursuit of creating highly efficient, multi-target drugs. This emphasis reflects the collaborative effort to advance pharmaceutical innovation with a focus on achieving optimal outcomes for patients.

3.2 Other factors of influencing hyperuricemia

Beyond the ongoing exploration of transporter protein inhibitors, various factors contribute to the modulation of transporter protein activity, influencing serum uric acid levels within the human body.

Lifestyle adjustments, such as dietary modifications and weight loss through exercise, have been identified as effective strategies to reduce serum uric acid levels and mitigate the risk of gout. However, it is crucial to note that abrupt weight loss can induce ketosis, a process that, via the URAT1, enhances uric acid reabsorption, consequently elevating serum uric acid levels [111]. Abdominal obesity-induced insulin resistance and hyperinsulinemia exacerbate uric acid reabsorption, facilitated by the sodium and monocarboxylate transporter (SMCT) exchanging with URAT1, leading to an upsurge in serum uric acid levels.

Advertisement

4. Conclusion

Hyperuricemia is a metabolic syndrome characterized by a persistently high serum uric acid level, primarily due to inadequate uric acid excretion by the kidneys and excessive reabsorption. The excretion and reabsorption of uric acid are important processes in uric acid metabolism, with several transport proteins, particularly URAT1, GLUT9, and ABCG2, found in the proximal tubules of the kidneys and intestines, playing a crucial role in the transmembrane transport of large-molecule urate salts and contributing significantly to the balance of serum uric acid.

The process of transporting uric acid by transport proteins is complex, involving exchanges of energy, potential, and surrounding substances. The deeper mechanisms of this process require further exploration. Transport proteins serve as crucial hubs in uric acid metabolism, and inhibitors targeting these proteins, particularly those involved in reabsorption, have long been one of the most important therapeutic approaches for managing hyperuricemia. Marketed drugs like probenecid, benzbromarone, sulfinpyrazone, and lesinurad effectively impede transporter proteins but may elicit varying degrees of adverse reactions in the body. New agents with better efficacy and safety are on the way for development.

Presently, the field is witnessing the development and modification of numerous compounds and natural products. Striking a balance between enhancing drug specificity and ensuring safety, particularly in drugs targeting critical transporter proteins, is paramount. As more novel modalities have been successfully delivered as new drugs, we would like to see whether the new modalities targeting transporters will benefit more patients. Additionally, the exploration of unknown uric acid transporter proteins and addressing other factors influencing these proteins represent the industry’s concerted efforts to elevate drug effectiveness in the treatment of hyperuricemia.

Advertisement

Acknowledgments

We appreciate the assistance and support provided by the Nanjing Municipal Food and Drug Supervision and Testing Institute for this article.

References

  1. 1. Jalal DI et al. Uric acid as a target of therapy in CKD. American Journal of Kidney Diseases. 2013;61(1):134-146
  2. 2. Jiang Q , Han M, Tang XP, Luo CG, Gong X. Guidelines for diagnosis and treatment of gout and hyperuricemia. Journal of Traditional Chinese Medicine. 2021;62(14):1276-1288
  3. 3. Johnson RJ et al. Hyperuricemia, acute and chronic kidney disease, hypertension, and cardiovascular disease: Report of a scientific workshop organized by the National Kidney Foundation. American Journal of Kidney Diseases. 2018;71(6):851-865
  4. 4. Chen-Xu M et al. Contemporary prevalence of gout and hyperuricemia in the United States and decadal trends: The National Health and nutrition examination survey, 2007-2016. Arthritis & Rhematology. 2019;71(6):991-999
  5. 5. Nutmakul T. A review on benefits of quercetin in hyperuricemia and gouty arthritis. Saudi Pharmaceutical Journal. 2022;30(7):918-926
  6. 6. Li L, Zhang Y, Zeng C. Update on the epidemiology, genetics, and therapeutic options of hyperuricemia. American Journal of Translational Research. 2020;12(7):3167-3181
  7. 7. Zhang WZ. Why does hyperuricemia not necessarily induce gout? Biomolecules. 2021;11(2):280
  8. 8. Braga F et al. Hyperuricemia as risk factor for coronary heart disease incidence and mortality in the general population: A systematic review and meta-analysis. Clinical Chemistry and Laboratory Medicine. 2016;54(1):7-15
  9. 9. Si K et al. Hyperuricemia and the risk of heart failure: Pathophysiology and therapeutic implications. Frontiers in Endocrinology (Lausanne). 2021;12:770815
  10. 10. Jiang J et al. Prevalence of diabetes in patients with hyperuricemia and gout: A systematic review and meta-analysis. Current Diabetes Reports. 2023;23(6):103-117
  11. 11. Waheed Y, Yang F, Sun D. Role of asymptomatic hyperuricemia in the progression of chronic kidney disease and cardiovascular disease. The Korean Journal of Internal Medicine. 2021;36(6):1281-1293
  12. 12. Ichida K et al. Decreased extra-renal urate excretion is a common cause of hyperuricemia. Nature Communications. 2012;3:764
  13. 13. Yanai H et al. Molecular biological and clinical understanding of the pathophysiology and treatments of hyperuricemia and its association with metabolic syndrome, cardiovascular diseases and chronic kidney disease. International Journal of Molecular Sciences. 2021;22(17):9221
  14. 14. So A, Thorens B. Uric acid transport and disease. The Journal of Clinical Investigation. 2010;120(6):1791-1799
  15. 15. Sorensen LB. Role of the intestinal tract in the elimination of uric acid. Arthritis and Rheumatism. 1965;8(5):694-706
  16. 16. Yinhui L, Ming L, Lili DUAN, Jieli YUAN. Research progress on the relationship between intestinal flora disturbance and hyperuricemia. Chinese Journal of Microecology. 2023;35(02):229-233
  17. 17. Giacomini KM et al. Membrane transporters in drug development. Nature Reviews Drug Discovery. 2010;9(3):215-236
  18. 18. Hediger MA et al. The ABCs of solute carriers: Physiological, pathological and therapeutic implications of human membrane transport proteins: Introduction. Pflügers Archiv. 2004;447(5):465-468
  19. 19. Gyimesi G, Hediger MA. Transporter-mediated drug delivery. Molecules. 2023;28:3
  20. 20. Moore JM et al. ABC transporters: Human disease and pharmacotherapeutic potential. Trends in Molecular Medicine. 2023;29(2):152-172
  21. 21. Oswald S. Organic anion transporting polypeptide (OATP) transporter expression, localization and function in the human intestine. Pharmacology & Therapeutics. 2019;195:39-53
  22. 22. Nigam SK, Granados JC. OAT, OATP, and MRP drug transporters and the remote sensing and Signaling theory. Annual Review of Pharmacology and Toxicology. 2023;63:637-660
  23. 23. Salhab A et al. Sodium(+)/taurocholate cotransporting polypeptide as target therapy for liver fibrosis. Gut. 2022;71(7):1373-1385
  24. 24. Nigam SK et al. The organic anion transporter (OAT) family: A systems biology perspective. Physiological Reviews. 2015;95(1):83-123
  25. 25. Nies AT, Schaeffeler E, Schwab M. Hepatic solute carrier transporters and drug therapy: Regulation of expression and impact of genetic variation. Pharmacology & Therapeutics. 2022;238:108268
  26. 26. Koepsell H. Organic cation transporters in health and disease. Pharmacological Reviews. 2020;72(1):253-319
  27. 27. Tan PK et al. Coevolution of URAT1 and uricase during primate evolution: Implications for serum urate homeostasis and gout. Molecular Biology and Evolution. 2016;33(9):2193-2200
  28. 28. Klaassen CD, Aleksunes LM. Xenobiotic, bile acid, and cholesterol transporters: Function and regulation. Pharmacological Reviews. 2010;62(1):1-96
  29. 29. Beaudoin JJ, Brouwer KLR, Malinen MM. Novel insights into the organic solute transporter alpha/beta, OSTα/β: From the bench to the bedside. Pharmacology & Therapeutics. 2020;211:107542
  30. 30. Adibi SA. The oligopeptide transporter (Pept-1) in human intestine: Biology and function. Gastroenterology. 1997;113(1):332-340
  31. 31. Dawed AY et al. Variation in the plasma membrane monoamine transporter (PMAT) (encoded by SLC29A4) and organic cation transporter 1 (OCT1) (encoded by SLC22A1) and gastrointestinal intolerance to metformin in type 2 diabetes: An IMI DIRECT study. Diabetes Care. 2019;42(6):1027-1033
  32. 32. Kobayashi M et al. Transport function, regulation, and biology of human monocarboxylate transporter 1 (hMCT1) and 4 (hMCT4). Pharmacology & Therapeutics. 2021;226:107862
  33. 33. Fang Y et al. Monocarboxylate transporter 4 inhibition potentiates hepatocellular carcinoma immunotherapy through enhancing T cell infiltration and immune attack. Hepatology. 2023;77(1):109-123
  34. 34. Iametti S, Bonomi F, Di Nunzio M. Dietary polyphenols and In vitro intestinal fructose uptake and transport: A systematic literature review. International Journal of Molecular Sciences. 2022;23(22):14355
  35. 35. Robey RW et al. Revisiting the role of ABC transporters in multidrug-resistant cancer. Nature Reviews. Cancer. 2018;18(7):452-464
  36. 36. Szatmári P, Ducza E. Changes in expression and function of placental and intestinal P-gp and BCRP transporters during pregnancy. International Journal of Molecular Sciences. 2023;24(17):13089
  37. 37. Soroka CJ, Boyer JL. Biosynthesis and trafficking of the bile salt export pump, BSEP: Therapeutic implications of BSEP mutations. Molecular Aspects of Medicine. 2014;37:3-14
  38. 38. Nigam SK. The SLC22 transporter family: A paradigm for the impact of drug transporters on metabolic pathways, Signaling, and disease. Annual Review of Pharmacology and Toxicology. 2018;58:663-687
  39. 39. Chang SY et al. Liver and kidney on chips: Microphysiological models to understand transporter function. Clinical Pharmacology and Therapeutics. 2016;100(5):464-478
  40. 40. Faber KN, Müller M, Jansen PL. Drug transport proteins in the liver. Advanced Drug Delivery Reviews. 2003;55(1):107-124
  41. 41. Kalliokoski A, Niemi M. Impact of OATP transporters on pharmacokinetics. British Journal of Pharmacology. 2009;158(3):693-705
  42. 42. Ballatori N et al. OST alpha-OST beta: A key membrane transporter of bile acids and conjugated steroids. Frontiers in Bioscience (Landmark Ed). 2009;14(8):2829-2844
  43. 43. Koepsell H. Glucose transporters in the small intestine in health and disease. Pflügers Archiv. 2020;472(9):1207-1248
  44. 44. Estudante M et al. Intestinal drug transporters: An overview. Advanced Drug Delivery Reviews. 2013;65(10):1340-1356
  45. 45. Andrade Sierra J, Flores Fonseca MM. Renal handling of uric acid. Contributions to Nephrology. 2018;192:1-7
  46. 46. Esparza Martín N, García Nieto V. Hypouricemia and tubular transport of uric acid. Nefrología. 2011;31(1):44-50
  47. 47. Mandal AK et al. Uricosuric targets of tranilast. Pharmacology Research & Perspectives. 2017;5(2):e00291
  48. 48. Hosomi A et al. Extra-renal elimination of uric acid via intestinal efflux transporter BCRP/ABCG2. PLoS One. 2012;7(2):e30456
  49. 49. Sarkadi B et al. ABCG2 – A transporter for all seasons. FEBS Letters. 2004;567(1):116-120
  50. 50. Woodward OM. ABCG2: The molecular mechanisms of urate secretion and gout. American Journal of Physiology. Renal Physiology. 2015;309(6):F485-F488
  51. 51. Matsuo H, Chiba T, Nagamori S, Nakayama A, Domoto H, Phetdee K, et al. Mutations in glucose transporter 9 gene SLC2A9 cause renal hypouricemia. American Journal of Human Genetics. 2008;83(6):744-751
  52. 52. Hoque KM et al. The ABCG2 Q141K hyperuricemia and gout associated variant illuminates the physiology of human urate excretion. Nature Communications. 2020;11(1):2767
  53. 53. Dalbeth N et al. Gout. Lancet. 2021;397(10287):1843-1855
  54. 54. Xu L et al. Recent advances on uric acid transporters. Oncotarget. 2017;8(59):100852-100862
  55. 55. Hosoyamada M et al. Function and localization of urate transporter 1 in mouse kidney. Journal of the American Society of Nephrology. 2004;15(2):261-268
  56. 56. Enomoto A et al. Molecular identification of a renal urate anion exchanger that regulates blood urate levels. Nature. 2002;417(6887):447-452
  57. 57. Sato M et al. Identification and functional characterization of uric acid transporter Urat1 (Slc22a12) in rats. Biochimica et Biophysica Acta. 2011;1808(6):1441-1447
  58. 58. Gopal E et al. Cloning and functional characterization of human SMCT2 (SLC5A12) and expression pattern of the transporter in kidney. Biochimica et Biophysica Acta. 2007;1768(11):2690-2697
  59. 59. Shin HJ et al. Interactions of urate transporter URAT1 in human kidney with uricosuric drugs. Nephrology (Carlton). 2011;16(2):156-162
  60. 60. Nakayama A et al. Genetic epidemiological analysis of hypouricaemia from 4993 Japanese on non-functional variants of URAT1/SLC22A12 gene. Rheumatology (Oxford). 2022;61(3):1276-1281
  61. 61. Eraly SA et al. Multiple organic anion transporters contribute to net renal excretion of uric acid. Physiological Genomics. 2008;33(2):180-192
  62. 62. 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. 2016;35(10-12):543-549
  63. 63. Fadieieva A et al. Role of SLC2A9 and ABCG2 gene polymorphisms in origin of hyperuricemia and gout. Georgian Medical News. 2016;252:79-83
  64. 64. Doege H et al. Activity and genomic organization of human glucose transporter 9 (GLUT9), a novel member of the family of sugar-transport facilitators predominantly expressed in brain and leucocytes. The Biochemical Journal. 2000;350(Pt 3):771-776
  65. 65. Anzai N et al. Recent advances in renal urate transport: Characterization of candidate transporters indicated by genome-wide association studies. Clinical and Experimental Nephrology. 2012;16(1):89-95
  66. 66. Caulfield MJ et al. SLC2A9 is a high-capacity urate transporter in humans. PLoS Medicine. 2008;5(10):e197
  67. 67. Kimura T et al. Expression of SLC2A9 isoforms in the kidney and their localization in polarized epithelial cells. PLoS One. 2014;9(1):e84996
  68. 68. Anzai N et al. Plasma urate level is directly regulated by a voltage-driven urate efflux transporter URATv1 (SLC2A9) in humans. The Journal of Biological Chemistry. 2008;283(40):26834-26838
  69. 69. Bibert S et al. Mouse GLUT9: evidences for a urate uniporter. American Journal of Physiology. Renal Physiology. 2009;297(3):F612-F619
  70. 70. Preitner F et al. Glut9 is a major regulator of urate homeostasis and its genetic inactivation induces hyperuricosuria and urate nephropathy. Proceedings of the National Academy of Sciences of the United States of America. 2009;106(36):15501-15506
  71. 71. Huls M et al. The breast cancer resistance protein transporter ABCG2 is expressed in the human kidney proximal tubule apical membrane. Kidney International. 2008;73(2):220-225
  72. 72. Ohashi Y et al. Evaluation of ABCG2-mediated extra-renal urate excretion in hemodialysis patients. Scientific Reports. 2023;13(1):93
  73. 73. Yano H et al. Uric acid transporter ABCG2 is increased in the intestine of the 5/6 nephrectomy rat model of chronic kidney disease. Clinical and Experimental Nephrology. 2014;18(1):50-55
  74. 74. Eckenstaler R, Benndorf RA. The role of ABCG2 in the pathogenesis of primary hyperuricemia and gout-an update. International Journal of Molecular Sciences. 2021;22(13):6678
  75. 75. Sarkadi B, Homolya L, Hegedűs T. The ABCG2/BCRP transporter and its variants - from structure to pathology. FEBS Letters. 2020;594(23):4012-4034
  76. 76. Cunningham R et al. Sodium-hydrogen exchanger regulatory factor-1 interacts with mouse urate transporter 1 to regulate renal proximal tubule uric acid transport. Journal of the American Society of Nephrology. 2007;18(5):1419-1425
  77. 77. Mandal AK, Mount DB. The molecular physiology of uric acid homeostasis. Annual Review of Physiology. 2015;77:323-345
  78. 78. Witkowska K et al. Human SLC2A9a and SLC2A9b isoforms mediate electrogenic transport of urate with different characteristics in the presence of hexoses. American Journal of Physiology. Renal Physiology. 2012;303(4):F527-F539
  79. 79. Wang L, Sweet DH. Renal organic anion transporters (SLC22 family): Expression, regulation, roles in toxicity, and impact on injury and disease. The AAPS Journal. 2013;15(1):53-69
  80. 80. Merriman TR, Dalbeth N. The genetic basis of hyperuricaemia and gout. Joint, Bone, Spine. 2011;78(1):35-40
  81. 81. Van Aubel RA et al. Human organic anion transporter MRP4 (ABCC4) is an efflux pump for the purine end metabolite urate with multiple allosteric substrate binding sites. American Journal of Physiology. Renal Physiology. 2005;288(2):F327-F333
  82. 82. Sweet DH. Organic anion transporter (Slc22a) family members as mediators of toxicity. Toxicology and Applied Pharmacology. 2005;204(3):198-215
  83. 83. Bahn A et al. Identification of a new urate and high affinity nicotinate transporter, hOAT10 (SLC22A13). The Journal of Biological Chemistry. 2008;283(24):16332-16341
  84. 84. Thangaraju M et al. c/ebpdelta null mouse as a model for the double knock-out of slc5a8 and slc5a12 in kidney. The Journal of Biological Chemistry. 2006;281(37):26769-26773
  85. 85. Mori K et al. Kidney-specific expression of a novel mouse organic cation transporter-like protein. FEBS Letters. 1997;417(3):371-374
  86. 86. Eraly SA et al. The molecular pharmacology of organic anion transporters: From DNA to FDA. Molecular Pharmacology. 2004;65(3):479-487
  87. 87. Eraly SA, Monte JC, Nigam SK. Novel slc22 transporter homologs in fly, worm, and human clarify the phylogeny of organic anion and cation transporters. Physiological Genomics. 2004;18(1):12-24
  88. 88. Robbins N et al. The history and future of probenecid. Cardiovascular Toxicology. 2012;12(1):1-9
  89. 89. Lee MH et al. A benefit-risk assessment of benzbromarone in the treatment of gout. Was its withdrawal from the market in the best interest of patients? Drug Safety. 2008;31(8):643-665
  90. 90. Miner JN et al. Lesinurad, a novel, oral compound for gout, acts to decrease serum uric acid through inhibition of urate transporters in the kidney. Arthritis Research & Therapy. 2016;18(1):214
  91. 91. Yu H et al. Safety and efficacy of benzbromarone and febuxostat in hyperuricemia patients with chronic kidney disease: A prospective pilot study. Clinical and Experimental Nephrology. 2018;22(6):1324-1330
  92. 92. Pui K, Gow PJ, Dalbeth N. Efficacy and tolerability of probenecid as urate-lowering therapy in gout; clinical experience in high-prevalence population. The Journal of Rheumatology. 2013;40(6):872-876
  93. 93. Hautekeete ML et al. Severe hepatotoxicity related to benzarone: A report of three cases with two fatalities. Liver. 1995;15(1):25-29
  94. 94. Tausche AK et al. Lesinurad monotherapy in gout patients intolerant to a xanthine oxidase inhibitor: A 6 month phase 3 clinical trial and extension study. Rheumatology (Oxford). 2017;56(12):2170-2178
  95. 95. Hoy SM. Lesinurad: First global approval. Drugs. 2016;76(4):509-516
  96. 96. Uda J et al. Discovery of Dotinurad (FYU-981), a new phenol derivative with highly potent uric acid lowering activity. ACS Medicinal Chemistry Letters. 2020;11(10):2017-2023
  97. 97. Tan PK et al. Discovery and characterization of verinurad, a potent and specific inhibitor of URAT1 for the treatment of hyperuricemia and gout. Scientific Reports. 2017;7(1):665
  98. 98. Toyoda Y et al. Inhibitory effect of citrus flavonoids on the in vitro transport activity of human urate transporter 1 (URAT1/SLC22A12), a renal re-absorber of urate. npj Science of Food. 2020;4:3
  99. 99. Chen L et al. The efficacy and mechanism of Chinese herbal medicines in lowering serum uric acid levels: A systematic review. Frontiers in Pharmacology. 2020;11:578318
  100. 100. Jiang LL et al. Bioactive compounds from plant-based functional foods: A promising choice for the prevention and management of hyperuricemia. Food. 2020;9(8):973
  101. 101. Zhong L et al. Oxyberberrubine, a novel liver microsomes-mediated secondary metabolite of berberine, alleviates hyperuricemic nephropathy in mice. Phytomedicine. 2023;108:154521
  102. 102. Chang YH et al. Anti-inflammatory and anti-Hyperuricemic effects of chrysin on a high fructose corn syrup-induced hyperuricemia rat model via the amelioration of urate transporters and inhibition of NLRP3 inflammasome Signaling pathway. Antioxidants (Basel). 2021;10(4):564
  103. 103. Ai G et al. Hypouricemic and nephroprotective effects of palmatine from cortex Phellodendri Amurensis: A uric acid modulator targeting Keap1-Nrf2/NLRP3 axis. Journal of Ethnopharmacology. 2023;301:115775
  104. 104. Li Y et al. Apigenin ameliorates hyperuricemic nephropathy by inhibiting URAT1 and GLUT9 and relieving renal fibrosis via the Wnt/β-catenin pathway. Phytomedicine. 2021;87:153585
  105. 105. Hu Q et al. (E)-2-(4-bromophenyl)-1-(2, 4-dihydroxyphenyl)ethanone oxime is a potential therapeutic agent for treatment of hyperuricemia through its dual inhibitory effects on XOD and URAT1. Biomedicine & Pharmacotherapy. 2017;86:88-94
  106. 106. Zhao T et al. Novel human urate transporter 1 inhibitors as hypouricemic drug candidates with Favorable Druggability. Journal of Medicinal Chemistry. 2020;63(19):10829-10854
  107. 107. Zhao ZA et al. CDER167, a dual inhibitor of URAT1 and GLUT9, is a novel and potent uricosuric candidate for the treatment of hyperuricemia. Acta Pharmacologica Sinica. 2022;43(1):121-132
  108. 108. Warrell Jr RP, Piwinski JJ, Acquist LLC. Bifunctional compounds and use for reducing uric acid levels. American patent US10093631B2. 2018. p. 45
  109. 109. Gao XNX, Huang YLH. Engineering red blood cells for treating gout and hyperuricemia diseases. Westlake therapeutics Hangzhou Co. Ltd. Chinese patent WO2021083278A1. 2021. p. 43
  110. 110. Gao X, Nie X, Ren J, et al. Modified red blood cells and uses thereof for treating hyperuricemia and gout. Westlake therapeutics Hangzhou Co. Ltd. Chinese patent WO2022166913A1. 2022. p. 35
  111. 111. Kakutani-Hatayama M et al. Nonpharmacological Management of Gout and Hyperuricemia: Hints for better lifestyle. American Journal of Lifestyle Medicine. 2017;11(4):321-329

Written By

Siqi Zhang, Jingwen Wang, Shuangxiang Wang, Zekai Dai, Lei Zhang and Feng Xue

Reviewed: 25 April 2024 Published: 22 May 2024