Next Article in Journal
Anti-Melanogenic Activity of Ethanolic Extract from Garcinia atroviridis Fruits Using In Vitro Experiments, Network Pharmacology, Molecular Docking, and Molecular Dynamics Simulation
Previous Article in Journal
Bioactive Compounds and Antioxidant Capacity of Pulp, Peel and Seeds from Jeriva (Syagrus romanzoffiana)
Previous Article in Special Issue
Gender Influence on XOR Activities and Related Pathologies: A Narrative Review
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Emerging Roles of Xanthine Oxidoreductase in Chronic Kidney Disease

Department of Medicine, Division of Nephrology, The Icahn School of Medicine at Mount Sinai, One Gustave Levy Place, Box 1243, New York, NY 10029, USA
*
Author to whom correspondence should be addressed.
Antioxidants 2024, 13(6), 712; https://doi.org/10.3390/antiox13060712
Submission received: 3 May 2024 / Revised: 9 June 2024 / Accepted: 10 June 2024 / Published: 12 June 2024

Abstract

:
Xanthine Oxidoreductase (XOR) is a ubiquitous, essential enzyme responsible for the terminal steps of purine catabolism, ultimately producing uric acid that is eliminated by the kidneys. XOR is also a physiological source of superoxide ion, hydrogen peroxide, and nitric oxide, which can function as second messengers in the activation of various physiological pathways, as well as contribute to the development and the progression of chronic conditions including kidney diseases, which are increasing in prevalence worldwide. XOR activity can promote oxidative distress, endothelial dysfunction, and inflammation through the biological effects of reactive oxygen species; nitric oxide and uric acid are the major products of XOR activity. However, the complex relationship of these reactions in disease settings has long been debated, and the environmental influences and genetics remain largely unknown. In this review, we give an overview of the biochemistry, biology, environmental, and current clinical impact of XOR in the kidney. Finally, we highlight recent genetic studies linking XOR and risk for kidney disease, igniting enthusiasm for future biomarker development and novel therapeutic approaches targeting XOR.

1. Introduction

Xanthine oxidoreductase (XOR) is the rate-limiting enzyme in the terminal steps of purine metabolism, which is the process of generating the building blocks of DNA and RNA. XOR’s reaction results in the production of uric acid (UA) in humans, of which two-thirds are excreted by the kidneys into urine, while the remaining one-third is eliminated via the gut. XOR can also produce reactive oxygen species (ROS) as it oxidizes hypoxanthine to xanthine as well as xanthine to UA [1,2,3]. XOR activity can be influenced by environmental factors, such as purine-rich and Western diets, infections, and increasing temperatures. Various disease states such as diabetes and aging can also enhance XOR activity [4,5]. Consequently, hyperuricemia (defined as elevated levels of serum UA) together with the generation of excess ROS can elicit damage to tissues, particularly on the endothelium [6,7,8,9,10].
XOR-derived ROS provides protection against infection, however, excess ROS through this pathway can also lead to inflammation and the development of oxidative distress-related tissue injury [11]. XOR-derived UA is considered a causal metabolite in the development of gout and a risk factor in the development of chronic kidney disease (CKD) and cardiovascular disease [12,13,14,15,16]. The presence of XOR in the blood can be used as a biomarker for a number of conditions, hence, XOR has been a drug target of interest for nearly 70 years and XOR inhibitors, either purine mimetics or selective non-purine drugs, have been effective at lowering serum UA levels [11,15,17,18,19,20,21,22]. However, these drugs have many limitations to their long-term use. Recent studies have found a correlation between genetic variants that regulate XOR activity and susceptibility to organ damage, suggesting a significant role of XOR and its products in this process [5,23]. In this review, we summarize the biochemical, physiological, and clinical implications of enhanced XOR activity and highlight the mechanistic implications of endothelial activation and ROS interplay in CKD.

2. XOR Biochemistry

XOR catalyzes the oxidation of hypoxanthine to xanthine and xanthine to UA whilst concomitantly reducing NAD+ to NADH as a dehydrogenase (xanthine dehydrogenase; XDH) or generates ROS, superoxide (O2•−) and hydrogen peroxide (H2O2) in its oxidase (xanthine oxidase; XO) form [10,11]. XOR is a molybdo-flavoenzyme and exists as a homodimer, with each subunit comprising an essential molybdenum cofactor (MoCo), two iron-sulfur clusters ([2Fe-2S] centers), and a flavin adenine dinucleotide (FAD) moiety where NAD+ binds [24]. The MoCo center facilitates substrate-level oxidation via water, and the subsequent electron transfer through the [2Fe-2S] centers to FAD, which in the case of XO, transfers the electrons to molecular oxygen producing O2•− and H2O2 [25,26]. Cofactor availability also can impact the activity of XOR, particularly by the presence of NAD+, which competes with molecular oxygen at the FAD site of XDH [25]. XDH oxidizes NAD+ and generates H+ from water forming NADH [Figure 1, below]. XDH requires ~500µM NAD+ to generate uric acid at its maximal activity, whereas XO produces UA independent of physiological NAD+ levels, underscoring the competitiveness of the FAD site between NAD+ (Kd = 25 µM) and O2 [25,27]. Moreover, studies have demonstrated the conserved local sulfhydryl groups of XDH, Cys535, Cys992, Cys1316, and Cys1324 play an effective role in regulating the post-translational conversion of XDH to XO, ultimately determining the sensitivity of the FAD site for O2 binding and subsequent generation of ROS [28]. This conversion from XDH to XO through oxidation is reversible; however, under prolonged ischemic conditions and diabetes, the conversion occurs irreversibly via proteolysis [29,30]. UA is the endpoint of purine metabolism in humans and some primates, while allantoin is the endpoint in most mammals [31]. UA is oxidized by uricase (UOX) to form allantoin. Allantoin results from a ring-opening oxidation of the purine skeleton at the C-2 position, which improves water solubility. Animals with UOX improve renal elimination of products of purine catabolism compared to humans who lack UOX which can accumulate pathological levels of UA [32].
UOX was once responsible for further oxidizing UA in early hominids, producing a much more water-soluble (i.e., readily dissolves in urine for excretion relative to UA) allantoin [Figure 1, below]. UOX has been pseudogenized in humans and was previously active in the human peroxisome where XOR has been documented to subcellularly localize, like it does in rat and taurus livers [31,33,34]. Generation of H2O2, under these circumstances, would have restricted effusion from the peroxisome and maintained the redox state through catalase activity, the enzyme responsible for decomposing H2O2 to water and O2 [35,36]. In humans the kidney plays a critical role in the regulation of serum UA levels through the reabsorption of filtered UA, as well as the secretion of urate via urate transporters [37]. Thus, without UOX, XOR production of UA must be well-regulated to prevent the pathological accumulation of UA.

3. XOR Biology: Products and Functions

Surplus or deficiency in XOR can result in increased or decreased serum UA levels (hyperuricemia or hypouricemia, respectively) and are associated with several prevalent diseases including hypertension, CKD, and hereditary xanthinuria [38,39,40,41]. Xanthinuria is a rare genetic disorder causing the accumulation of xanthine in blood and urine, which can lead to xanthine urolithiasis or kidney stones and subsequent renal failure [42]. Mutations in exon 16 in XDH result in xanthinuria, limiting the function of XOR and therefore hindering the oxidation of xanthine, leading to the accumulation of xanthine crystals in the kidney. The low levels of UA (i.e., hypouricemia) can also lead to renal damage causing the accumulation of purines, and an imbalance of the redox state promoting oxidative distress [43,44,45]. On the other hand, hyperuricemia has also been linked to renal pathogeneses, likely through increased UA and/or through the excess generation of ROS in tissues, including endothelial damage and dysfunction by decreasing endothelial NO availability [43,44,46].

3.1. XOR Activity and Cell Damage

The role of XOR and its products in mediating cell damage has been previously described to occur via several mechanisms. Studies have shown that following entry into the cell, XOR exerts prooxidant effects through the increased production of reactive species such as O2•−, H2O2, UA and isoprostanes [47,48,49]. It can also activate NADPH oxidase (NOX) and endothelin-1 expression [50,51,52]. In endothelial cells, this would result in the activation and promotion of pro-inflammatory activity due to increased vascular endothelial permeability and local vasodilation. It has been also demonstrated that endothelium-bound XOR can inhibit NO-dependent cyclic guanosine monophosphate production in smooth muscle cells, therefore, contributing to impaired vasorelaxation states [53].
Mitochondria play a preeminent role in cellular metabolic processes such as oxidative phosphorylation, during which O2•− is also produced [44]. Hyperuricemia has been shown to mediate damage in endothelial cells by activating the mitochondrial sodium-calcium exchanger (NCXMito), which results in mitochondrial calcium overload and the increased production of O2•− [54]. UA-mediated alterations in mitochondrial morphology and reduced intracellular ATP production have also been implicated in endothelial dysfunction [55]. Studies by Huang et al. have shown that one mechanism through which UA stimulates ROS production and mediates endothelial cell dysfunction is via the activation of aldose reductase [56]. Aldose reductase is an important enzyme of the polyol pathway that catalyzes the conversion of glucose to sorbitol, which is a process that generates large amounts of intracellular ROS [57]. The major consequences of UA-mediated ROS production include cellular DNA damage, protein damage, and lipid peroxidation [51,58,59], which can lead to inactivation of intracellular enzymes, activation of maladaptive responses that promote proinflammatory signaling cascades, and cell death [59,60].
The kidney consists of a heterogeneous population of cells that carry out several energy-demanding physiological processes. Indeed, mitochondrial content and oxygen consumption of renal cells such as tubular epithelial cells (TEC) are amongst the highest in the body [61,62,63]. The implication of this is that interference with cellular metabolic processes can be pathogenic to TECs and interstitial cells, as well as cells within the glomerular compartment including podocytes, glomerular endothelial cells, and mesangial cells. UA in hyperuricemia stimulates epithelial-to-mesenchymal transition (EMT) in tubules and activates profibrotic signaling pathways and proteins like TGFβ-SMAD3 and MMPs [64,65,66]. UA has also been shown to stimulate the TLR4/NFκB signaling pathway which is involved in renal inflammation and fibrosis [64]. In the vasculature, XOR induces endothelial dysfunction via various mechanisms, including increased ROS and inhibition of endothelial nitric oxide synthase (eNOS) expression and nitric oxide (NO) release [Figure 2, below]. Additionally, UA induces endothelial-to-mesenchymal transition, mesangial cell proliferation through NOX/ROS/ERK signaling, and the increased expression of profibrotic markers in mesangial cells such as TGFβ, α-SMA, and fibronectin [67,68,69,70]. These mechanisms contribute to glomerulosclerosis and the breakdown of the glomerular filtration barrier resulting in proteinuria and progressive disease [71].
In oxidative distress states, such as those seen in diabetes, XOR activity inhibition was shown to attenuate oxidative distress and protect against diabetic kidney injury by inhibiting the vascular endothelial growth factor (VEGF)-NOX signaling pathway in human glomerular endothelial cells [72]. The study showed that increased XOR in STZ-induced diabetic kidney disease was associated with increased VEGF/VEGFR1 and VEGFR3 levels in the kidneys, followed by the activation of NOX1, NOX2, and NOX4 expression, also by FoxO3a phosphorylation and the activation of eNOS. Other studies demonstrated that XOR inhibition can prevent mitochondrial oxidative DNA damage in endothelial cells, which is characteristic of early injury [5,73]. It has also been suggested by some authors that XOR can be endocytosed by endothelial cells, thus inducing endothelial dysfunction; however, more investigations are required to validate such a mechanism [74]. In all, a potential feed-forward interplay exists between XOR-derived ROS enhancing mitochondrial-derived ROS release and endothelial cell stress [Figure 2, below], which could provide a mechanism that would promote a vicious cycle of sustained stress and accumulation of oxidized products within the microvasculature.

3.2. Role of XOR in CKD

Dysregulation of XOR activity has been implicated in the pathogenesis of various cardiorenal and metabolic disease states. In the context of CKD, XOR contributes to both the development and progression through direct and indirect mechanisms. Hyperuricemia due to increased XOR activity can result in the increased deposition of poorly soluble urate crystals in the renal tubules and medullary interstitial space [75]. This can lead to gouty nephropathy, which is characterized by urate kidney stones and tubular injury and can ultimately lead to renal failure [76,77]. Preclinical and clinical studies have demonstrated that hyperuricemia contributes to CKD via activation of the Nod-like receptor protein 3 (NLRP3) inflammasome-mediated inflammation and downstream pro-inflammatory events such as increased renal expression of IL1β and IL18 with increased macrophage activation and infiltration [78,79,80]. Other reports have shown that XOR-mediated renal inflammation is indeed an important pathological mechanism for the development of glomerular and tubular injury, and renal fibrosis seen in AKI and CKD possibly through the action of XOR-derived ROS [67,81,82,83,84,85,86]. Additionally, hyperuricemia can activate the intrarenal renin-angiotensin-aldosterone system (RAAS), thereby resulting in vasoconstriction of glomerular arterioles leading to glomerular ischemia, glomerular endothelial dysfunction, glomerulosclerosis, and renal fibrosis [47,87,88].
In obese diabetic mice, increased XOR activity was shown to be correlated with adipose tissue inflammation, and pharmacological blockade of XOR rescued markers of metabolic syndrome in these mice [89]. Clinical studies have established a positive correlation between hyperuricemia and markers of metabolic syndrome in young adults and in patients with asymptomatic hyperuricemia [90,91]. Importantly, treatment with allopurinol reduced these markers in patients with hyperuricemia. Increased XOR activity and hyperuricemia can promote hypertension through various mechanisms described above such as activating inflammation, decreasing endothelial NO availability, mediating endothelial dysfunction, activating the RAAS, and vascular remodeling [92]. These reports support that the mechanisms mediated by XOR activity are also involved in the development of diabetes and hypertension, which are the leading risk factors for CKD.
The apical surface of the endothelial cells is covered by the negatively charged endothelial glycocalyx, a hydrated gel-like structure consisting of glycoproteins and proteoglycans covalently linked to the glycosaminoglycans (GAG), heparan sulfate, chondroitin sulfate, and hyaluronic acid [93]. The release of XOR into circulation from organs such as the liver promotes the accumulation of XO in peripheral endothelia by binding to GAGs at the plasma membrane [8]. The XO binding to GAGs in human endothelial cells has been demonstrated with a Kd of 6 nM, which is extremely favorable and underscores the potential for consequences to high XO accumulation and activity [8]. Once bound to endothelial GAGs, XO can mediate endothelial cell damage by inhibiting NO production through uncoupling eNOS and impact the vascular tone critical for the development of hypertension [38,72,94,95,96]. This reaction can also result in the production of peroxynitrite (ONOO-), an extremely potent reactive species that can do extensive damage, by nitrating protein residues on tyrosine leading to functional impairment of cells or cell death [97]. The glomerular endothelial glycocalyx rich in GAGs has been suggested to serve as a vulnerable site for XOR accumulation in diabetic mice [Figure 2, above], and shedding of the glycocalyx in the glomerulus would result in XO release, promoting endothelial dysfunction and glomerular injury and proteinuria [7,98].
Indeed, type 2 diabetes (T2D) and XOR activity have been suggested to play a role in diabetic kidney disease (DKD) pathogenesis [39,90,99,100]. Oxidative distress induced by XOR was also shown to be critical in the development of DKD, through ROS-mediated cellular damage and activation of cell death pathways [5,44]. Importantly, in humans, XO and UA were reported to be independent predictors of albuminuria, where an elevation in 1 µmol/L UA or 1U/L of XO increases albuminuria by 1.5% [101]. Mouse and rat studies have explored the effects of XOR inhibition in T2D, and showed anti-inflammatory effects, reduced distress, and reduced UA levels [102]. However, recent metanalyses in humans found that administration of allopurinol in DKD did not significantly improve renal function nor albuminuria, yet another metanalysis showed a significant reduction in UA levels with the use of allopurinol and preserved renal function in addition to reducing proteinuria [103,104]. Also, outcomes from clinical studies concluded that 3 years of sustained reductions of blood levels of UA with allopurinol did not benefit diabetic patients with mild to moderate kidney disease [105,106]. The studies’ conclusions have been disputed by the community because both studies included large numbers of patients with normal serum UA levels, and therefore they did not test whether treating hyperuricemia was beneficial, but rather tested whether lowering UA would be beneficial, and it is well recognized that normal UA levels do not increase the risk for CKD [107]. It should be noted that allopurinol while lowering UA, can cause excess ROS production through its recognized action of self-oxidation to form oxypurinol, ultimately leading to electron transfer to FAD, resulting in the reduction of O2 and more ROS formation [108,109,110]. Other reports have shown that allopurinol has off-target consequences, inhibiting other enzymes in the purine and pyrimidine metabolic pathways, which can also lead to kidney damage [111]. Non-purine XOR inhibitors have been shown to inhibit XOR-derived ROS damage and renal injury with greater selectivity than purine mimetics [112,113]. Febuxostat is one such drug, and although its use has had controversial outcomes, meta-analyses show the benefit of febuxostat in CKD and its superiority over allopurinol [113,114,115,116,117,118]. Randomized controlled trials involving 65 patients with hyperuricemia concomitant with diabetic nephropathy demonstrated that topiroxostat, another non-purine XOR inhibitor decreased serum uric acid and prevented a decline in renal function, without improving albuminuria in patients with diabetes [119], while in some other randomized controlled trials, topiroxostat displayed antialbuminuric effects [120,121]. The differences in observed effects of XOR inhibition may be attributed to the sample size of these clinical studies, duration of treatment, stage of patients’ CKD, levels of uric acid, or the drug effects as described above. In summary, although mixed results have been observed from pre-clinical to clinical studies with XOR inhibitors in diabetic and non-diabetic CKD summarized in Table 1 below, there is substantial evidence for the role of XOR in CKD pathophysiology and the beneficial effects for XOR inhibition in CKD.
Therapeutic targeting of XOR activity may, in fact, benefit a subgroup of patients [107,133]. This is supported by reports demonstrating genetic variations in XOR are significantly associated with CKD in a cohort of hypertensives [134]. While XOR activity can play a consequential role in the development of kidney diseases, unrelated renal insults, such as polycystic kidney disease (PKD), an inherited disorder, have also been correlated to changes in XOR activity [135]. PKD is a common genetic disease present in 1:400 people and is driven by either autosomal dominant or recessive genes [136,137]. Described by the development of cysts in the kidney, PKD exhibits oxidative distress, insults to insulin signaling, hyperuricemia, and endothelial dysfunction, which are linked to XOR activity in PKD [135,138,139]. In autosomal dominant PKD (ADPKD), hyperuricemia develops as renal function declines, underscoring the positive correlation with XOR activity as the disease progresses [140]. Interestingly, hyperuricemia in PKD has also been found to be a risk factor for nephromegaly and an earlier onset of hypertension [43]. Targeting XOR with febuxostat in PKD was shown to be beneficial by increasing patients’ eGFR, which supports the pathologic role of XOR activity in this condition [141]. Studies focused on the role of XOR activity in the development and progression of PKD have only recently begun, and an understanding of the mechanisms involved is just emerging. Future studies on how XOR and its products contribute to the onset and progression of PKD are warranted.

4. XOR Activity & Diet

While disease settings such as diabetes, hypertension, and PKD can influence XOR activity, environmental, and dietary factors may also mediate XOR activity [142,143]. XOR activity in driving hyperuricemia is greatly impacted by dietary patterns, such as purine-rich and western diets (WDs) [6,9,143]. Purine-rich diets consist of frequent intake of shellfish, organ meats, legumes, and alcohol [142,144,145]. Ingestion of purine-rich foods supplies the highly active purine salvage pathway, which is vital in producing nucleic acids for bioenergetic regulation, cellular replication, and sustaining transcriptional and translational activities through the generation of mRNA and tRNAs, and this has been elegantly demonstrated in model organisms such as E. coli and S. cerevisiae [146]. While de novo purine synthesis can occur, it is energetically expensive, whereas dietary sources of purines are abundant and can be modified with much less energy expenditure in the form of ATP [147]. Thus, dietary purine contents have been suggested to regulate UA production by XOR and form the basis for nutritional interventions in hyperuricemic diseases—predominantly gout [144,148]. While purine-rich diets directly contribute to XOR activity by means of supplying upstream substrate(s), WDs, notably high in saturated fats, fructose, and cholesterol, have also been associated with increased XOR expression and activity with hyperuricemia, gout, and metabolic diseases [149,150]. WDs have been implicated as major drivers of insulin resistance [151]. WDs, which often have subjects consuming nutrient-dense, ultra-processed foods more frequently, promote oxidative stress and metabolic disease [151,152,153]. Nitrites, a major mineral involved in food processing in WDs, directly impact XOR activity to reduce nitrites to NO [154,155]. This mechanism may act as a compensatory role for NO production by eNOS when XOR mediates the uncoupling of eNOS [156]. However, more investigation into the mechanism of WDs effect on XOR activity and its impact on health is needed.

4.1. Diet-Induced XOR Activity in CVD and Fatty Liver

Chronic WD exposure is a recognized and costly risk factor for cardiovascular disease (CVD), supported by numerous studies demonstrating that WD can increase cardiac distress, inflammation, and cell death [157,158,159,160]. WD has been shown to also alter cardiac contractility in rats as a result of dysregulated fatty acid metabolism [161]. Moreover, advanced glycation end products, which are markers for organ damage and inflammation, were found to accumulate in mice fed a WD, suggesting activation of receptors for advanced glycation end products, is consequential in WD-induced CVD [158]. In an ApoE knockout mouse model of atherosclerosis, dietary tungsten inhibited XO, and mitigated the development of atherosclerosis from WD, suggesting that dietary tungsten, could have a positive impact on WD-induced XOR activity [Table 2, below] [162]. Risks for cardiac insults in humans are increased during hyperuricemia, having shown that diets high in sodium, typical in some WDs, are associated with increased XO activity and left ventricular hypertrophy in resistant hypertension [15]. WDs directly impact hepatic health and the susceptibility to developing the metabolic-associated fatty liver disease (MAFLD) [163,164]. While predominantly produced and released by the liver, hepatic XOR plays a focal role in the progression of MAFLD [6,165]. WDs promote insulin resistance, which in turn drives the pathogenesis of MAFL and metabolic-associated steatohepatitis (MASH). In mice challenged with WD, XOR inhibition was able to attenuate insulin resistance and mitigate the progression of MASH [6,166]. Interestingly, a recent study revealed that endothelial-sourced released hypoxanthine was increased in vitro, suggesting that activated endothelia directly promotes XOR activity by providing metabolic substrates for XOR [Figure 2, above] [167]. Kawachi et al. treated HUVECs primed by human liver S9—a fraction of microsomal and cytosolic enzymes from the liver, which are rich with XOR—with the selective XOR inhibitor, topiroxostat, and found that hypoxanthine was still highly produced compared to attenuated levels of xanthine and UA [167]. This further highlights the negative impact of XOR on endothelial function as well as the complex relationship between XOR regulation, endothelial activation, and dysfunction, and its contribution to CVD and MAFLD pathogeneses [168].

4.2. Diet, XOR Activity in CKD

High fructose intake has been demonstrated to impact renal health and function in humans, increasing the risk for hyperuricemia and kidney stones [169]. Recent evidence in mice revealed that WD-induced MASH promoted the development of CKD, which can be mitigated by healthy liver transplantation, suggesting that a hepatorenal relationship is responsible for CKD progression in this setting [170]. Indeed, there is mounting research suggesting a strong causal role for fructose consumption on UA production; including a study in mice showing that UA can stimulate hepatic fructokinase in a seemingly vicious positive feedback loop promoting metabolic disease [4,171]. Likewise, dietary fructose in mice has been shown to induce de novo purine anabolism, which can strain downstream purine catabolism by XOR [143]. Indeed, recent studies employing transcriptomics and metabolomics in livers from mice fed high fructose, underscore the causal relationship between fructose feeding and XOR activity, demonstrating significantly increased gene expression of purine metabolic enzymes, especially XOR, as well as elevated hypoxanthine, adenine, adenosine, AMP, and guanine metabolite levels [143]. Proximal tubules are responsible for fructose reabsorption for renal gluconeogenesis and are susceptible to high fructose diets resulting in tubulointerstitial injury, supporting the notion that high fructose feeding is associated with poor outcomes, promoting CKD pathogenesis [172,173,174]. In vitro studies have found that fructose administered to HK-2 cells promotes fructokinase activity and subsequently the production of proinflammatory factors [175]. In light of this evidence, strategies to mitigate fructose-induced renal damage and fibrosis are becoming increasingly important. Recent advances in glucagon-like peptide-1 (GLP-1) receptor agonists, such as the use of dulaglutide, have shown promising results in reducing renal fibrosis, in albino rats chronically fed high fructose [176]. Indeed, a study in Taiwan found that GLP-1 receptor agonists provide renal benefits in T2D patients in contrast to long-acting insulins [177]. Altogether, dietary patterns can play a role in mediating XOR activity and future research exploring nutritional interventions could be beneficial in managing or even mitigating chronic diseases like CKD [178].
Table 2. Factors regulating XOR and outcomes.
Table 2. Factors regulating XOR and outcomes.
FactorImpact on XOR
Expression/Activity
Biological EffectEffect SizeReference
Purine-rich DietIncreaseGreater UA; elevated UA precursors promote purine catabolic activity of XOROdds ratio: 1.024 for animal-derived foods[145]
Western DietIncreaseGreater UAOdds ratio: 2.15 for animal-derived and fried foods[179]
TungstenReduceReduced ROS; Prevents Molybdenum incorporation at the MoCo site thereby reducing XO-derived ROS production.N/A[180,181]
HeatIncreaseRenal function (measured in eGFR) declined with rising UA. Electrolyte hydration by sugarcane workers appeared preventative to the declineadj β = −10.4 and 8.1, respectively[182]
XanthinuriaNon-functionalReduced UA; elevated purines and formation of xanthine stones1:69,000 people (combined incidence of hereditary types I and II)[183]
XDH rs207454ReduceImproved survival for ‘C/C’ carriers facing gastric cancer compared to ‘A/A’ or ‘A/C’ carriers.Hazard Ratio: 1.53 for A/A and A/C carriers[184]
XDH rs185925IncreaseHigher XOR expression was associated with acute respiratory distress syndrome in septic African American patientsOdds ratio: 1.464 for C/T carriers[23]
XDH rs1884725IncreaseIncreased serum creatinine (a predictor for renal dysfunction) in septic African American patients.β = 0.504[23]
XDH rs4952085IncreaseIncreased serum creatinine (a predictor for renal dysfunction) in septic African American patients.β = 0.493[23]

5. XOR Activity and the Environment

5.1. XOR and Heavy Metal Exposures

Susceptibility to environmental impacts beyond diets on XOR activity and its products has also been explored. Studies have found that XORs are susceptible to dysregulation as a result of toxic metal intake, excessive heat exposure, and infectious diseases [23,80,185,186,187]. Heavy metal (e.g., lead, copper, tungsten, etc.) exposure can come from a variety of sources and has been positively associated, particularly lead, with the development of hyperuricemia in gout patients [188]. Interestingly, copper has also been suggested to play a role in dysregulating XOR activity and may have a greater impact on people compared to lead, due to its wide dietary availability [185]. On the other hand, dietary tungsten has been used to inhibit XOR activity and does so by displacing molybdenum due to similar bonding properties in the same periodic group [162,189]. More research focused on the impact of heavy metals on XOR activity could help better our understanding the impact of heavy metal exposure on human health and toxicity.

5.2. XOR and CKDu

With climate change causing increased temperatures, more and more people are being chronically exposed to extreme heat, which is associated with an increased risk of CKD [190,191]. A recent study in mice employed heat stress to significantly induce renal injury and found that treatment of allopurinol mitigated the fibrotic and inflammatory effects that were observed in heat-stressed mice, suggesting that XOR activity is triggered in chronic heat stress and may mediate kidney injury in this context [186].
Over the past decades, there has been an increase in the prevalence of CKD of unknown etiology (CKDu). First, reported amongst young rural farming communities in El Salvador, CKDu has since been reported in regions of the world such as India and Sri Lanka [192,193,194,195]. CKDu has also been reported among miners, and construction and transportation workers living in similar hotter and lower altitudes areas of the U.S. Pacific coast [196]. CKDu is typically asymptomatic, however, individuals exhibit increased serum creatinine with little or no proteinuria [192,197]. Additionally, kidney biopsies of affected individuals reveal tubulointerstitial disease which typically progresses to renal failure [198]. Since the first observation of CKDu was made, various pathophysiologic mechanisms for development have been proposed [193]. Based on studies over the years, Dr. Johnson and colleagues propose that the ‘chronic recurrent dehydration’ hypothesis is most plausible [182,196]. This hypothesis proposes that high levels of physical activity, with insufficient hydration in hot environments, lead to dehydration, increased serum osmolarity, and subclinical-grade muscle injury [Table 2, above]. Subclinical-grade muscle injury and rhabdomyolysis lead to the release of purines from damaged muscles, which eventually results in increased XOR and production of UA from these purine substrates. This, along with volume depletion due to dehydration leads to hyperuricemia. UA in the setting of hyperuricemia can cause urate crystal deposition in renal tubules and contribute to the acidification of urine, which together initiate events that lead to tubular injury [64,199]. Other reports support the common occurrence of hyperuricemia in individuals with CKDu [200,201]. In the quest to understand the cause of CKDu, other investigators undertook multi-omics approach by exposing Zebrafish to water from a CKDu-prevalent region of Sri Lanka [202]. The researchers identified necroptosis and purine metabolism pathways to be dysregulated in CKDu [202]. Although heat exposure, possible concomitant particulate matter, and toxicants exposure in these environments may play a role in promoting CKDu in those at risk, the role of XOR and UA in the pathogenesis of the disease may be important [203,204,205].

6. Genetic Regulation of XOR

While components of diet and the environment have some effect on XOR expression and activity, a recent metanalysis from ARIC, CARDIA, CHS, FHS, and NHANES III studies in the U.S., using 16,760 patients (8414 men and 8346) of European descent demonstrated that genetic factors, specifically single nucleotide polymorphisms (SNPs) in 30 different genes, play a systemic and consequential role in mediating serum UA [148]. The genetic loss of functional human XDH results in renal decline, which is reflected in a hereditary xanthinuria model of XDH loss in rats [Table 2, above] [42,45]. There also exists genetic polymorphisms in XDH (rs207454), which influence gastric cancer survival in Chinese patients and breast cancer survival among a Spanish cohort through increased XOR expression [184,206,207]. However, in certain scenarios, the regulation of the XOR gene could potentially contribute to disease progression in various settings. The human XOR is transcriptionally regulated by the presence of E-box and TATA-like elements in the promoter region (−258 to −1) of XDH [208]. In a study evaluating risk for sepsis and associated organ failures, six intronic variants enriched with regulatory elements for XOR were associated with risk of sepsis among African Americans (rs206805, rs513311, rs185925, rs561525, rs2163059, rs13387204) [Table 2, above]. Interestingly, the rs185925 variant was identified to influence XOR activity, and two common SNPs (rs1884725 and rs4952085) were in tight linkage disequilibrium which provided strong evidence for association with increased levels of serum creatinine and risk of renal dysfunction [23,184]. Conversely, the same study found that the missense variant rs17011368 was associated with enhanced mortality in septic European Americans with acute respiratory distress syndrome compared to African Americans [23]. Thus, variations in XDH regulation are associated with risk for several negative outcomes, and differences in ethnic risk profiles may begin to explain some of the health disparity (i.e., higher morbidity and mortality) in African American patients.
In line with the epidemiologic evidence from human studies, promoter polymorphisms of XOR have been associated with increased activity and gene expression, and increased XOR in circulation is strongly associated with ESRD [209,210]. In an unbiased integrative systems-genetics study in mice, our group identified the presence of a variation in the C/ebpβ binding element of XDH in DBA/2J mice with high risk for kidney injury phenotype, compared to resistant phenotype exhibited by C57BL/6J mice in the setting of diabetes [5,73]. By substituting the low-risk with high-risk variants from the DBA/2J strain using CRISPR Cas9, Wang et al. were able to generate a kidney injury susceptible C57BL/6J mouse model; B6-Xorem1, where B6-Xorem1 mice exhibited significantly higher levels of XOR activity with superimposed diabetes, high-fat, diet-induced metabolic syndrome and with aging [5]. Mechanistically, the study confirmed that these variants could influence the binding of transcription factor C/ebpβ. Although C/ebpβ is normally expressed at low levels in kidneys, this study detected higher C/ebpβ expression in glomeruli of diabetic DBA/2J and B6-Xorem1 mice but not in diabetic C57BL/6J. Primary GECs from B6-Xorem1 mice showed nuclear translocation of C/ebpβ with high glucose or oxidized LDL, while primary podocytes from diabetic C57BL/6J did not. RNA interference for C/ebpβ prevented the high glucose-induced XOR expression and ROS production, confirming its role in modulating XOR activity in a high glucose milieu. Furthermore, the study demonstrated that high-risk strains (DBA/2J and B6-Xorem1) had increased XOR activity with concomitant oxidative distress in endothelial cells, glomerular injury, and proteinuria, which could be rescued by febuxostat treatment. The data underscores the critical role of Xor regulation in influencing kidney injury in diabetes, which is also relevant to other diabetic complications and aging [211,212,213]. Importantly, XOR promoter orthologues were identified in humans having diabetic complications including kidney disease in the UK BioBank and in the Mount Sinai BioMe Biobank [5]. Identifying individuals at risk for developing DKD or other diabetic complications is crucial for early intervention, prevention of further complications, and improving overall health outcomes and quality of life for those individuals. From a mechanistic standpoint, this work advocated for a precision-medicine approach, in which genetic risk variants in the XOR promoter that result in increased activity could potentially guide treatment decisions. Future research will evaluate whether XOR promoter variants could be potential genetic biomarker predictors of progressive disease, aiding in the identification and treatment of those at risk.

7. Novel Therapeutic Approaches Targeting XOR

The use of current XOR inhibitors is associated with serious side effects that limit their use in patients. For instance, purine derivatives such as allopurinol have been associated with gastrointestinal distress, renal toxicity, rash, and eosinophilia [111,214]. Non-purine structured inhibitor febuxostat is associated with an increased risk of cardiovascular mortality and in 2017, it received a black box warning from the FDA [215]. Considering the established involvement of XOR in several diseases including kidney disease, there have been global efforts geared towards the development of effective pharmacological inhibitors of XOR that do not cause noteworthy adverse reactions. Optimism remains around tackling the aberrant purine metabolism in the development of ADPKD and DKD; XRx-008 and XRx-225, novel formulations of oxypurinol that have been designed to slow or reverse disease progression are currently in Phase II and Phase I clinical trials, respectively. Recently, candidate non-purine compounds, namely ALS-1, -8, -15, and -28, have been shown to effectively inhibit XO in a reversible manner [216]. These compounds may offer a safer therapeutic option for disorders that are characterized by a dysregulation in XOR activity. Other approaches aim to combine the inhibition of both XOR and URAT1 in order to reduce uric acid and avoid the issue of insufficient potency by single-target drugs [217]. Progress in this space over the last 20 years has been captured in a comprehensive review elsewhere, which highlights the use of novel drug design approaches such as fragment-based drug design or molecular hybridization, or the combination of both in designing a new class of XOR inhibitors that could potentially effectively eliminate side effects while achieving the desired potency and efficacy [218]. Interestingly, the development of new inhibitor candidates for XOR so far is largely based on derivatives inspired by existing drugs, however, this approach has resulted in limited success. Therefore, new approaches such as the use of exogenous UOX (i.e., enzyme replacement therapy) to lower UA levels could be further explored in the setting of gouty nephropathy [219]. Despite the challenges that are being encountered by the currently approved inhibitors of XOR, pharmacological targeting of XOR still holds enormous therapeutic potential, especially for chronic diseases that are associated with increased XOR activity in non-diabetic and diabetic kidney diseases. Exciting and new emerging trends in this research are becoming evident, as scientists are exploring natural compounds for XOR inhibition with pleiotropic effects, there are more developments for new tools for identifying drug candidates that target optimal efficacy and safety profiles, as well as efforts to identify the patients who may be most responsive.

8. Conclusions

In conclusion, XOR activity is complex in health and dysregulation can be induced by a myriad of environmental factors, such as diet, heat, toxic exposure(s), or diseases, or aging. It is important to note that there are sex differences in the expression of XOR in humans and animal models [220,221,222]. The sex-based differences can influence XOR expression and activity potentially leading to higher susceptibility to chronic diseases [221,223]. Considering XOR in CKD, there are promising results from current therapeutics at alleviating some key factors involved in CKD pathogenesis; however, to avoid off-target effects novel therapeutics should be more thoroughly investigated for clinical potential. Exciting new evidence on the genetics of XOR regulation on susceptibility to diseases, especially wherever oxidative damage is involved, is of particular importance. Genetic polymorphisms of XOR could serve as valuable biomarkers of susceptibility, aiding in the identification of patient sub-groups that would benefit from targeted therapies. Understanding the interaction of XOR and various influencing factors such as diseases, aging, diet, toxicant exposures, and climate change, all potentially leading to CKD [Figure 3, below], will assist in accurately diagnosing individuals at risk and providing them with more precise and effective treatments.

Author Contributions

Writing—original draft preparation, H.W.K., U.S.E. and I.S.D.; writing—review and editing, H.W.K., U.S.E. and I.S.D.; supervision, I.S.D.; project administration, I.S.D.; funding acquisition, I.S.D. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by National Institutes of Health Grants numbers R01DK097253 and R01DK139395, and the Department of Defense CDMRP grant E01 W81XWH-20-1-0836 (to I.S.D).

Acknowledgments

Illustrations were created using ChemDraw 22.2.0 and BioRender.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Bobulescu, I.A.; Moe, O.W. Renal Transport of Uric Acid: Evolving Concepts and Uncertainties. Adv. Chronic Kidney Dis. 2012, 19, 358–371. [Google Scholar] [CrossRef]
  2. Aliciguzel, Y.; Ozen, I.; Aslan, M.; Karayalcin, U. Activities of xanthine oxidoreductase and antioxidant enzymes in different tissues of diabetic rats. J. Lab. Clin. Med. 2003, 142, 172–177. [Google Scholar] [CrossRef]
  3. El Ridi, R.; Tallima, H. Physiological functions and pathogenic potential of uric acid: A review. J. Adv. Res. 2017, 8, 487–493. [Google Scholar] [CrossRef] [PubMed]
  4. Lanaspa, M.A.; Sanchez-Lozada, L.G.; Cicerchi, C.; Li, N.; Roncal-Jimenez, C.A.; Ishimoto, T.; Le, M.; Garcia, G.E.; Thomas, J.B.; Rivard, C.J.; et al. Uric Acid Stimulates Fructokinase and Accelerates Fructose Metabolism in the Development of Fatty Liver. PLoS ONE 2012, 7, e47948. [Google Scholar] [CrossRef]
  5. Wang, Q.; Qi, H.; Wu, Y.; Yu, L.; Bouchareb, R.; Li, S.; Lassén, E.; Casalena, G.; Stadler, K.; Ebefors, K.; et al. Genetic susceptibility to diabetic kidney disease is linked to promoter variants of XOR. Nat. Metab. 2023, 5, 607–625. [Google Scholar] [CrossRef]
  6. Nishikawa, T.; Nagata, N.; Shimakami, T.; Shirakura, T.; Matsui, C.; Ni, Y.; Zhuge, F.; Xu, L.; Chen, G.; Nagashimada, M.; et al. Xanthine oxidase inhibition attenuates insulin resistance and diet-induced steatohepatitis in mice. Sci. Rep. 2020, 10, 815. [Google Scholar] [CrossRef] [PubMed]
  7. Itano, S.; Kadoya, H.; Satoh, M.; Nakamura, T.; Murase, T.; Sasaki, T.; Kanwar, Y.S.; Kashihara, N. Non-purine selective xanthine oxidase inhibitor ameliorates glomerular endothelial injury in InsAkita diabetic mice. Am. J. Physiol. Physiol. 2020, 319, F765–F772. [Google Scholar] [CrossRef] [PubMed]
  8. Adachi, T.; Fukushima, T.; Usami, Y.; Hirano, K. Binding of human xanthine oxidase to sulphated glycosaminoglycans on the endothelial-cell surface. Biochem. J. 1993, 289, 523–527. [Google Scholar] [CrossRef]
  9. Yokose, C.; McCormick, N.; Choi, H.K. The role of diet in hyperuricemia and gout. Curr. Opin. Rheumatol. 2021, 33, 135–144. [Google Scholar] [CrossRef] [PubMed]
  10. Kelley, E.E.; Khoo, N.K.; Hundley, N.J.; Malik, U.Z.; Freeman, B.A.; Tarpey, M.M. Hydrogen peroxide is the major oxidant product of xanthine oxidase. Free. Radic. Biol. Med. 2009, 48, 493–498. [Google Scholar] [CrossRef]
  11. Bortolotti, M.; Polito, L.; Battelli, M.G.; Bolognesi, A. Xanthine oxidoreductase: One enzyme for multiple physiological tasks. Redox Biol. 2021, 41, 101882. [Google Scholar] [CrossRef] [PubMed]
  12. Duskin-Bitan, H.; Cohen, E.; Goldberg, E.; Shochat, T.; Levi, A.; Garty, M.; Krause, I. The Degree of Asymptomatic Hyperu-ricemia and the Risk of Gout. A Retrospective Analysis of a Large Cohort. Clin. Rheumatol. 2014, 33, 549–553. [Google Scholar] [CrossRef] [PubMed]
  13. Petta, S.; Cammà, C.; Cabibi, D.; Di Marco, V.; Craxì, A. Hyperuricemia is associated with histological liver damage in patients with non-alcoholic fatty liver disease. Aliment. Pharmacol. Ther. 2011, 34, 757–766. [Google Scholar] [CrossRef] [PubMed]
  14. Li, L.; Yang, C.; Zhao, Y.; Zeng, X.; Liu, F.; Fu, P. Is hyperuricemia an independent risk factor for new-onset chronic kidney disease?: A systematic review and meta-analysis based on observational cohort studies. BMC Nephrol. 2014, 15, 122. [Google Scholar] [CrossRef] [PubMed]
  15. Butts, B.; Calhoun, D.A.; Denney, T.S.; Lloyd, S.G.; Gupta, H.; Gaddam, K.K.; Aban, I.; Oparil, S.; Sanders, P.W.; Patel, R.; et al. Plasma xanthine oxidase activity is related to increased sodium and left ventricular hypertrophy in resistant hypertension. Free Radic. Biol. Med. 2019, 134, 343–349. [Google Scholar] [CrossRef] [PubMed]
  16. Taufiq, F.; Li, P.; Miake, J.; Hisatome, I. Hyperuricemia as a Risk Factor for Atrial Fibrillation Due to Soluble and Crystalized Uric Acid. Circ. Rep. 2019, 1, 469–473. [Google Scholar] [CrossRef] [PubMed]
  17. Elion, G.B.; Kovensky, A.; Hitchings, G.H.; Metz, E.; Rundles, R. Metabolic studies of allopurinol, an inhibitor of xanthine oxidase. Biochem. Pharmacol. 1966, 15, 863–880. [Google Scholar] [CrossRef] [PubMed]
  18. Cicero, A.F.; Fogacci, F.; Cincione, R.I.; Tocci, G.; Borghi, C. Clinical Effects of Xanthine Oxidase Inhibitors in Hyperuricemic Patients. Med. Princ. Pract. 2021, 30, 122–130. [Google Scholar] [CrossRef] [PubMed]
  19. Pacher, P.; Nivorozhkin, A.; Szabó, C. Therapeutic Effects of Xanthine Oxidase Inhibitors: Renaissance Half a Century after the Discovery of Allopurinol. Pharmacol. Rev. 2006, 58, 87–114. [Google Scholar] [CrossRef] [PubMed]
  20. Miesel, R.; Zuber, M. Elevated levels of xanthine oxidase in serum of patients with inflammatory and autoimmune rheumatic diseases. Inflammation 1993, 17, 551–561. [Google Scholar] [CrossRef]
  21. Murase, T.; Oka, M.; Nampei, M.; Miyachi, A.; Nakamura, T. A highly sensitive assay for xanthine oxidoreductase activity using a combination of [13C2,15N2]xanthine and liquid chromatography/triple quadrupole mass spectrometry. J. Label. Compd. Radiopharm. 2016, 59, 214–220. [Google Scholar] [CrossRef] [PubMed]
  22. Kawachi, Y.; Fujishima, Y.; Nishizawa, H.; Nagao, H.; Nakamura, T.; Akari, S.; Murase, T.; Taya, N.; Omori, K.; Miyake, A.; et al. Plasma xanthine oxidoreductase activity in Japanese patients with type 2 diabetes across hospitalized treatment. J. Diabetes Investig. 2021, 12, 1512–1520. [Google Scholar] [CrossRef] [PubMed]
  23. Gao, L.; Rafaels, N.; Dudenkov, T.M.; Damarla, M.; Damico, R.; Maloney, J.P.; Moss, M.; Martin, G.S.; Sevransky, J.; Shanholtz, C.; et al. Xanthine oxidoreductase gene polymorphisms are associated with high risk of sepsis and organ failure. Respir. Res. 2023, 24, 177. [Google Scholar] [CrossRef] [PubMed]
  24. Olson, J.S.; Ballou, D.P.; Palmer, G.; Massey, V. The Mechanism of Action of Xanthine Oxidase. J. Biol. Chem. 1974, 249, 4363–4382. [Google Scholar] [CrossRef] [PubMed]
  25. Harris, C.M.; Massey, V. The Oxidative Half-reaction of Xanthine Dehydrogenase with NAD; Reaction Kinetics and Steady-state Mechanism. J. Biol. Chem. 1997, 272, 28335–28341. [Google Scholar] [CrossRef] [PubMed]
  26. Xia, M.; Dempski, R.; Hille, R. The Reductive Half-reaction of Xanthine Oxidase Reaction with Aldehyde Substrates and Identification of the Catalytically Labile Oxygen*. J. Biol. Chem. 1999, 274, 3323–3330. [Google Scholar] [CrossRef] [PubMed]
  27. Lee, M.-C.; Velayutham, M.; Komatsu, T.; Hille, R.; Zweier, J.L. Measurement and Characterization of Superoxide Generation from Xanthine Dehydrogenase: A Redox-Regulated Pathway of Radical Generation in Ischemic Tissues. Biochemistry 2014, 53, 6615–6623. [Google Scholar] [CrossRef] [PubMed]
  28. Nishino, T.; Okamoto, K.; Kawaguchi, Y.; Hori, H.; Matsumura, T.; Eger, B.T.; Pai, E.F.; Nishino, T. Mechanism of the Conversion of Xanthine Dehydrogenase to Xanthine Oxidase: Identification of the Two Cysteine Disulfide Bonds and Crystal Structure of a Non-Convertible Rat Liver Xanthine Dehydrogenase Mutant. J. Biol. Chem. 2005, 280, 24888–24894. [Google Scholar] [CrossRef] [PubMed]
  29. McKelvey, T.G.; Hollwarth, M.E.; Granger, D.N.; Engerson, T.D.; Landler, U.; Jones, H.P. Mechanisms of conversion of xanthine dehydrogenase to xanthine oxidase in ischemic rat liver and kidney. Am. J. Physiol. Liver Physiol. 1988, 254, G753–G760. [Google Scholar] [CrossRef]
  30. Weinstein, A.L.; Lalezarzadeh, F.D.; Soares, M.A.; Saadeh, P.B.; Ceradini, D.J. Normalizing dysfunctional purine metabolism accelerates diabetic wound healing. Wound Repair Regen. 2015, 23, 14–21. [Google Scholar] [CrossRef]
  31. Kratzer, J.T.; Lanaspa, M.A.; Murphy, M.N.; Cicerchi, C.; Graves, C.L.; Tipton, P.A.; Ortlund, E.A.; Johnson, R.J.; Gaucher, E.A. Evolutionary history and metabolic insights of ancient mammalian uricases. Proc. Natl. Acad. Sci. USA 2014, 111, 3763–3768. [Google Scholar] [CrossRef] [PubMed]
  32. Roman, Y.M. The Role of Uric Acid in Human Health: Insights from the Uricase Gene. J. Pers. Med. 2023, 13, 1409. [Google Scholar] [CrossRef] [PubMed]
  33. Angermüller, S.; Bruder, G.; Völkl, A.; Wesch, H.; Fahimi, H.D. Localization of Xanthine Oxidase in Crystalline Cores of Pe-roxisomes. A Cytochemical and Biochemical Study. Eur. J. Cell Biol. 1987, 45, 137–144. [Google Scholar] [PubMed]
  34. Frederiks, W.M.; Vreeling-Sindelárová, H. Ultrastructural localization of xanthine oxidoreductase activity in isolated rat liver cells. Acta Histochem. 2002, 104, 29–37. [Google Scholar] [CrossRef]
  35. Williams, J. The Decomposition of Hydrogen Peroxide by Liver Catalase. J. Gen. Physiol. 1928, 11, 309–337. [Google Scholar] [CrossRef] [PubMed]
  36. Veenhuis, M.; Bonga, S.E.W. Cytochemical localization of catalase and several hydrogen peroxide-producing oxidases in the nucleoids and matrix of rat liver peroxisomes. Histochem. J. 1979, 11, 561–572. [Google Scholar] [CrossRef] [PubMed]
  37. Schlesinger, N.; Pérez-Ruiz, F.; Lioté, F. Mechanisms and rationale for uricase use in patients with gout. Nat. Rev. Rheumatol. 2023, 19, 640–649. [Google Scholar] [CrossRef] [PubMed]
  38. Khosla, U.M.; Zharikov, S.; Finch, J.L.; Nakagawa, T.; Roncal, C.; Mu, W.; Krotova, K.; Block, E.R.; Prabhakar, S.; Johnson, R.J. Hyperuricemia induces endothelial dysfunction. Kidney Int. 2005, 67, 1739–1742. [Google Scholar] [CrossRef] [PubMed]
  39. Jalal, D.I.; Maahs, D.M.; Hovind, P.; Nakagawa, T. Uric Acid as a Mediator of Diabetic Nephropathy. Semin. Nephrol. 2011, 31, 459–465. [Google Scholar] [CrossRef]
  40. Kim, I.Y.; Lee, D.W.; Lee, S.B.; Kwak, I.S. The Role of Uric Acid in Kidney Fibrosis: Experimental Evidences for the Causal Relationship. BioMed Res. Int. 2014, 2014, 638732. [Google Scholar] [CrossRef]
  41. Sebesta, I.; Stiburkova, B.; Krijt, J. Hereditary xanthinuria is not so rare disorder of purine metabolism. Nucleosides Nucleotides Nucleic Acids 2018, 37, 324–328. [Google Scholar] [CrossRef] [PubMed]
  42. Gonçalves, P.L.; Diniz, H.; Tavares, I.; Dória, S.; Dong, J.; Kyriss, M.; Fairbanks, L.; Oliveira, J.P. Kidney Failure Secondary to Hereditary Xanthinuria due to a Homozygous Deletion of the XDH Gene in the Absence of Overt Kidney Stone Disease. Nephron 2024, 1–6. [Google Scholar] [CrossRef] [PubMed]
  43. Helal, I.; McFann, K.; Reed, B.; Yan, X.-D.; Schrier, R.W.; Fick-Brosnahan, G.M. Serum uric acid, kidney volume and progression in autosomal-dominant polycystic kidney disease. Nephrol. Dial. Transplant. 2013, 28, 380–385. [Google Scholar] [CrossRef] [PubMed]
  44. Daehn, I.S.; Ekperikpe, U.S.; Stadler, K. Redox regulation in diabetic kidney disease. Am. J. Physiol. Physiol. 2023, 325, F135–F149. [Google Scholar] [CrossRef] [PubMed]
  45. Dissanayake, L.V.; Zietara, A.; Levchenko, V.; Spires, D.R.; Angulo, M.B.; El-Meanawy, A.; Geurts, A.M.; Dwinell, M.R.; Palygin, O.; Staruschenko, A. Lack of xanthine dehydrogenase leads to a remarkable renal decline in a novel hypouricemic rat model. iScience 2022, 25, 104887. [Google Scholar] [CrossRef] [PubMed]
  46. Laakso, J.; Mervaala, E.; Himberg, J.-J.; Teräväinen, T.-L.; Karppanen, H.; Vapaatalo, H.; Lapatto, R. Increased Kidney Xanthine Oxidoreductase Activity in Salt-Induced Experimental Hypertension. Hypertension 1998, 32, 902–906. [Google Scholar] [CrossRef] [PubMed]
  47. Yu, M.-A.; Sánchez-Lozada, L.G.; Johnson, R.J.; Kang, D.-H. Oxidative stress with an activation of the renin–angiotensin system in human vascular endothelial cells as a novel mechanism of uric acid-induced endothelial dysfunction. J. Hypertens. 2010, 28, 1234–1242. [Google Scholar] [CrossRef] [PubMed]
  48. Roumeliotis, S.; Roumeliotis, A.; Dounousi, E.; Eleftheriadis, T.; Liakopoulos, V. Dietary Antioxidant Supplements and Uric Acid in Chronic Kidney Disease: A Review. Nutrients 2019, 11, 1911. [Google Scholar] [CrossRef] [PubMed]
  49. Battelli, M.G.; Polito, L.; Bortolotti, M.; Bolognesi, A. Xanthine Oxidoreductase-Derived Reactive Species: Physiological and Pathological Effects. Oxidative Med. Cell. Longev. 2016, 2016, 3527579. [Google Scholar] [CrossRef]
  50. Kadowaki, D.; Sakaguchi, S.; Miyamoto, Y.; Taguchi, K.; Muraya, N.; Narita, Y.; Sato, K.; Chuang, V.T.G.; Maruyama, T.; Otagiri, M.; et al. Direct Radical Scavenging Activity of Benzbromarone Provides Beneficial Antioxidant Properties for Hyperuricemia Treatment. Biol. Pharm. Bull. 2015, 38, 487–492. [Google Scholar] [CrossRef]
  51. Verzola, D.; Ratto, E.; Villaggio, B.; Parodi, E.L.; Pontremoli, R.; Garibotto, G.; Viazzi, F. Uric Acid Promotes Apoptosis in Human Proximal Tubule Cells by Oxidative Stress and the Activation of NADPH Oxidase NOX 4. PLoS ONE 2014, 9, e115210. [Google Scholar] [CrossRef] [PubMed]
  52. Chao, H.-H.; Liu, J.-C.; Lin, J.-W.; Chen, C.-H.; Wu, C.-H.; Cheng, T.-H. Uric acid stimulates endothelin-1 gene expression associated with NADPH oxidase in human aortic smooth muscle cells. Acta Pharmacol. Sin. 2008, 29, 1301–1312. [Google Scholar] [CrossRef]
  53. Houston, M.; Estevez, A.; Chumley, P.; Aslan, M.; Marklund, S.; Parks, D.A.; Freeman, B.A. Binding of Xanthine Oxidase to Vascular Endothelium: Kinetic Characterization and Oxidative Impairment of Nitric Oxide-Dependent Signaling. J. Biol. Chem. 1999, 274, 4985–4994. [Google Scholar] [CrossRef]
  54. Hong, Q.; Qi, K.; Feng, Z.; Huang, Z.; Cui, S.; Wang, L.; Fu, B.; Ding, R.; Yang, J.; Chen, X.; et al. Hyperuricemia induces endothelial dysfunction via mitochondrial Na+/Ca2+ exchanger-mediated mitochondrial calcium overload. Cell Calcium 2012, 51, 402–410. [Google Scholar] [CrossRef]
  55. Sánchez-Lozada, L.G.; Lanaspa, M.A.; Cristóbal-García, M.; García-Arroyo, F.; Soto, V.; Cruz-Robles, D.; Nakagawa, T.; Yu, M.A.; Kang, D.-H.; Johnson, R.J. Uric Acid-Induced Endothelial Dysfunction Is Associated with Mitochondrial Alterations and Decreased Intracellular ATP Concentrations. Nephron Exp. Nephrol. 2012, 121, e71–e78. [Google Scholar] [CrossRef] [PubMed]
  56. Huang, Z.; Hong, Q.; Zhang, X.; Xiao, W.; Wang, L.; Cui, S.; Feng, Z.; Lv, Y.; Cai, G.; Chen, X.; et al. Aldose reductase mediates endothelial cell dysfunction induced by high uric acid concentrations. Cell Commun. Signal. 2017, 15, 3. [Google Scholar] [CrossRef]
  57. Tang, W.H.; Martin, K.A.; Hwa, J. Aldose Reductase, Oxidative Stress, and Diabetic Mellitus. Front. Pharmacol. 2012, 3, 87. [Google Scholar] [CrossRef]
  58. Yang, L.; Chang, B.; Guo, Y.; Wu, X.; Liu, L. The role of oxidative stress-mediated apoptosis in the pathogenesis of uric acid nephropathy. Ren. Fail. 2019, 41, 616–622. [Google Scholar] [CrossRef] [PubMed]
  59. Kono, H.; Chen, C.-J.; Ontiveros, F.; Rock, K.L. Uric acid promotes an acute inflammatory response to sterile cell death in mice. J. Clin. Investig. 2010, 120, 1939–1949. [Google Scholar] [CrossRef]
  60. Daehn, I.; Karran, P. Immune Effector Cells Produce Lethal DNA Damage in Cells Treated with a Thiopurine. Cancer Res. 2009, 69, 2393–2399. [Google Scholar] [CrossRef]
  61. Tian, Z.; Liang, M. Renal metabolism and hypertension. Nat. Commun. 2021, 12, 963. [Google Scholar] [CrossRef] [PubMed]
  62. Bhargava, P.; Schnellmann, R.G. Mitochondrial energetics in the kidney. Nat. Rev. Nephrol. 2017, 13, 629–646. [Google Scholar] [CrossRef] [PubMed]
  63. Fuchs, M.A.; Wolf, M. Renal proximal tubule cells: Power and finesse. J. Clin. Investig. 2023, 133. [Google Scholar] [CrossRef] [PubMed]
  64. Zhou, Y.; Fang, L.; Jiang, L.; Wen, P.; Cao, H.; He, W.; Dai, C.; Yang, J. Uric Acid Induces Renal Inflammation via Activating Tubular NF-κB Signaling Pathway. PLoS ONE 2012, 7, e39738. [Google Scholar] [CrossRef] [PubMed]
  65. Kim, S.-M.; Choi, Y.-W.; Seok, H.-Y.; Jeong, K.-H.; Lee, S.-H.; Lee, T.-W.; Ihm, C.-G.; Lim, S.J.; Moon, J.-Y. Reducing Serum Uric Acid Attenuates TGF-β1-Induced Profibrogenic Progression in Type 2 Diabetic Nephropathy. Nephron Exp. Nephrol. 2012, 121, e109–e121. [Google Scholar] [CrossRef] [PubMed]
  66. Tao, M.; Shi, Y.; Tang, L.; Wang, Y.; Fang, L.; Jiang, W.; Lin, T.; Qiu, A.; Zhuang, S.; Liu, N. Blockade of ERK1/2 by U0126 alleviates uric acid-induced EMT and tubular cell injury in rats with hyperuricemic nephropathy. Am. J. Physiol. Physiol. 2019, 316, F660–F673. [Google Scholar] [CrossRef] [PubMed]
  67. Li, P.; Zhang, L.; Zhang, M.; Zhou, C.; Lin, N. Uric Acid Enhances PKC-Dependent ENOS Phosphorylationand Mediates Cellular ER Stress: A Mechanism for Uric Acid-Induced Endothelial Dysfunction. Int. J. Mol. Med. 2016, 37, 989–997. [Google Scholar] [CrossRef] [PubMed]
  68. Kang, D.H. Hyperuricemia and Progression of Chronic Kidney Disease: Role of Phenotype Transition of Renal Tubular and Endothelial Cells. Contrib. Nephrol. 2018, 192, 48–55. [Google Scholar] [CrossRef] [PubMed]
  69. Zhuang, Y.; Feng, Q.; Ding, G.; Zhao, M.; Che, R.; Bai, M.; Bao, H.; Zhang, A.; Huang, S. Activation of ERK1/2 by NADPH oxidase-originated reactive oxygen species mediates uric acid-induced mesangial cell proliferation. Am. J. Physiol. Physiol. 2014, 307, F396–F406. [Google Scholar] [CrossRef]
  70. Li, S.; Zhao, F.; Cheng, S.; Wang, X.; Hao, Y. Uric acid-induced endoplasmic reticulum stress triggers phenotypic change in rat glomerular mesangial cells. Nephrology 2013, 18, 682–689. [Google Scholar] [CrossRef]
  71. Daehn, I.S.; Duffield, J.S. The glomerular filtration barrier: A structural target for novel kidney therapies. Nat. Rev. Drug Discov. 2021, 20, 770–788. [Google Scholar] [CrossRef]
  72. Yang, K.-J.; Choi, W.J.; Chang, Y.-K.; Park, C.W.; Kim, S.Y.; Hong, Y.A. Inhibition of Xanthine Oxidase Protects against Diabetic Kidney Disease through the Amelioration of Oxidative Stress via VEGF/VEGFR Axis and NOX-FoxO3a-eNOS Signaling Pathway. Int. J. Mol. Sci. 2023, 24, 3807. [Google Scholar] [CrossRef]
  73. Qi, H.; Casalena, G.; Shi, S.; Yu, L.; Ebefors, K.; Sun, Y.; Zhang, W.; D’agati, V.; Schlondorff, D.; Haraldsson, B.; et al. Glomerular Endothelial Mitochondrial Dysfunction Is Essential and Characteristic of Diabetic Kidney Disease Susceptibility. Diabetes 2016, 66, 763–778. [Google Scholar] [CrossRef] [PubMed]
  74. Yoshida, S.; Kurajoh, M.; Fukumoto, S.; Murase, T.; Nakamura, T.; Yoshida, H.; Hirata, K.; Inaba, M.; Emoto, M. Association of plasma xanthine oxidoreductase activity with blood pressure affected by oxidative stress level: MedCity21 health examination registry. Sci. Rep. 2020, 10, 4437. [Google Scholar] [CrossRef]
  75. Maejima, I.; Takahashi, A.; Omori, H.; Kimura, T.; Takabatake, Y.; Saitoh, T.; Yamamoto, A.; Hamasaki, M.; Noda, T.; Isaka, Y.; et al. Autophagy sequesters damaged lysosomes to control lysosomal biogenesis and kidney injury. EMBO J. 2013, 32, 2336–2347. [Google Scholar] [CrossRef]
  76. Lusco, M.A.; Fogo, A.B.; Najafian, B.; Alpers, C.E. AJKD Atlas of Renal Pathology: Gouty Nephropathy. Am. J. Kidney Dis. 2017, 69, e5–e6. [Google Scholar] [CrossRef] [PubMed]
  77. Cao, L.; Zhao, T.; Xie, C.; Zheng, S.; Wan, W.; Zou, H.; Zhu, X. Performance of Ultrasound in the Clinical Evaluation of Gout and Hyperuricemia. J. Immunol. Res. 2021, 2021, 5550626. [Google Scholar] [CrossRef]
  78. Wu, M.; Ma, Y.; Chen, X.; Liang, N.; Qu, S.; Chen, H. Hyperuricemia causes kidney damage by promoting autophagy and NLRP3-mediated inflammation in rats with urate oxidase deficiency. Dis. Model. Mech. 2021, 14, dmm048041. [Google Scholar] [CrossRef] [PubMed]
  79. Zhang, Y.; Sui, X.; Xu, Y.; Gu, F.; Zhang, A.; Chen, J. Association between Nod-like receptor protein 3 inflammasome and gouty nephropathy. Exp. Ther. Med. 2020, 20, 195–204. [Google Scholar] [CrossRef]
  80. Ives, A.; Nomura, J.; Martinon, F.; Roger, T.; LeRoy, D.; Miner, J.N.; Simon, G.; Busso, N.; So, A. Xanthine oxidoreductase regulates macrophage IL1β secretion upon NLRP3 inflammasome activation. Nat. Commun. 2015, 6, 6555. [Google Scholar] [CrossRef]
  81. Andrade-Oliveira, V.; Foresto-Neto, O.; Watanabe, I.K.M.; Zatz, R.; Câmara, N.O.S. Inflammation in Renal Diseases: New and Old Players. Front. Pharmacol. 2019, 10, 1192. [Google Scholar] [CrossRef] [PubMed]
  82. Nomura, J.; Kobayashi, T.; So, A.; Busso, N. Febuxostat, a Xanthine Oxidoreductase Inhibitor, Decreases NLRP3-dependent Inflammation in Macrophages by Activating the Purine Salvage Pathway and Restoring Cellular Bioenergetics. Sci. Rep. 2019, 9, 17314. [Google Scholar] [CrossRef] [PubMed]
  83. Ohtsubo, T.; Matsumura, K.; Sakagami, K.; Fujii, K.; Tsuruya, K.; Noguchi, H.; Rovira, I.I.; Finkel, T.; Iida, M. Xanthine Oxidoreductase Depletion Induces Renal Interstitial Fibrosis Through Aberrant Lipid and Purine Accumulation in Renal Tubules. Hypertension 2009, 54, 868–876. [Google Scholar] [CrossRef] [PubMed]
  84. Ko, J.; Kang, H.; Kim, D.; Kim, M.; Ryu, E.; Lee, S.; Ryu, J.; Roncal, C.; Johnson, R.J.; Kang, D. Uric acid induced the phenotype transition of vascular endothelial cells via induction of oxidative stress and glycocalyx shedding. FASEB J. 2019, 33, 13334–13345. [Google Scholar] [CrossRef] [PubMed]
  85. Susztak, K.; Raff, A.C.; Schiffer, M.; Böttinger, E.P. Glucose-Induced Reactive Oxygen Species Cause Apoptosis of Podocytes and Podocyte Depletion at the Onset of Diabetic Nephropathy. Diabetes 2006, 55, 225–233. [Google Scholar] [CrossRef] [PubMed]
  86. Charlton, A.; Garzarella, J.; Jandeleit-Dahm, K.A.M.; Jha, J.C. Oxidative Stress and Inflammation in Renal and Cardiovascular Complications of Diabetes. Biology 2020, 10, 18. [Google Scholar] [CrossRef] [PubMed]
  87. Johnson, R.J.; Nakagawa, T.; Jalal, D.; Sánchez-Lozada, L.G.; Kang, D.-H.; Ritz, E. Uric acid and chronic kidney disease: Which is chasing which? Nephrol. Dial. Transplant. 2013, 28, 2221–2228. [Google Scholar] [CrossRef] [PubMed]
  88. Fan, S.; Zhang, P.; Wang, A.Y.; Wang, X.; Wang, L.; Li, G.; Hong, D. Hyperuricemia and its related histopathological features on renal biopsy. BMC Nephrol. 2019, 20, 95. [Google Scholar] [CrossRef] [PubMed]
  89. Baldwin, W.; McRae, S.; Marek, G.; Wymer, D.; Pannu, V.; Baylis, C.; Johnson, R.J.; Sautin, Y.Y. Hyperuricemia as a Mediator of the Proinflammatory Endocrine Imbalance in the Adipose Tissue in a Murine Model of the Metabolic Syndrome. Diabetes 2011, 60, 1258–1269. [Google Scholar] [CrossRef]
  90. Washio, K.; Kusunoki, Y.; Murase, T.; Nakamura, T.; Osugi, K.; Ohigashi, M.; Sukenaga, T.; Ochi, F.; Matsuo, T.; Katsuno, T.; et al. Xanthine oxidoreductase activity is correlated with insulin resistance and subclinical inflammation in young humans. Metabolism 2017, 70, 51–56. [Google Scholar] [CrossRef]
  91. Takir, M.; Kostek, O.; Ozkok, A.; Elcioglu, O.C.; Bakan, A.; Erek, A.; Mutlu, H.H.; Telci, O.; Semerci, A.; Odabas, A.R.; et al. Lowering Uric Acid with Allopurinol Improves Insulin Resistance and Systemic Inflammation in Asymptomatic Hyperuricemia. J. Investig. Med. 2015, 63, 924–929. [Google Scholar] [CrossRef] [PubMed]
  92. Kuwabara, M. Hyperuricemia, Cardiovascular Disease, and Hypertension. Pulse 2016, 3, 242–252. [Google Scholar] [CrossRef]
  93. Weinbaum, S.; Cancel, L.M.; Fu, B.M.; Tarbell, J.M. The Glycocalyx and Its Role in Vascular Physiology and Vascular Related Diseases. Cardiovasc. Eng. Technol. 2021, 12, 37–71. [Google Scholar] [CrossRef]
  94. Kanbay, M.; Yilmaz, M.I.; Sonmez, A.; Turgut, F.; Saglam, M.; Cakir, E.; Yenicesu, M.; Covic, A.; Jalal, D.; Johnson, R.J. Serum Uric Acid Level and Endothelial Dysfunction in Patients with Nondiabetic Chronic Kidney Disease. Am. J. Nephrol. 2011, 33, 298–304. [Google Scholar] [CrossRef]
  95. Idigo, W.O.; Reilly, S.; Zhang, M.H.; Zhang, Y.H.; Jayaram, R.; Carnicer, R.; Crabtree, M.J.; Balligand, J.-L.; Casadei, B. Regulation of Endothelial Nitric-oxide Synthase (NOS) S-Glutathionylation by Neuronal NOS Evidence of a Functional Interaction between Myocardial Constitutive Nos Isoforms *. J. Biol. Chem. 2012, 287, 43665–43673. [Google Scholar] [CrossRef] [PubMed]
  96. Li, Q.; Youn, J.-Y.; Cai, H. Mechanisms and consequences of ENOS dysfunction in hypertension. J. Hypertens. 2015, 33, 1128–1136. [Google Scholar] [CrossRef] [PubMed]
  97. Pacher, P.; Szabo, C. Role of the Peroxynitrite-Poly(ADP-Ribose) Polymerase Pathway in Human Disease. Am. J. Pathol. 2008, 173, 2–13. [Google Scholar] [CrossRef]
  98. Ebefors, K.; Wiener, R.J.; Yu, L.; Azeloglu, E.U.; Yi, Z.; Jia, F.; Zhang, W.; Baron, M.H.; He, J.C.; Haraldsson, B.; et al. Endothelin receptor-A mediates degradation of the glomerular endothelial surface layer via pathologic crosstalk between activated podocytes and glomerular endothelial cells. Kidney Int. 2019, 96, 957–970. [Google Scholar] [CrossRef]
  99. Miric, D.J.; Kisic, B.M.; Filipovic-Danic, S.; Grbic, R.; Dragojevic, I.; Miric, M.B.; Puhalo-Sladoje, D. Xanthine Oxidase Activity in Type 2 Diabetes Mellitus Patients with and without Diabetic Peripheral Neuropathy. J. Diabetes Res. 2016, 2016, 4370490. [Google Scholar] [CrossRef]
  100. Dehghan, A.; van Hoek, M.; Sijbrands, E.J.; Hofman, A.; Witteman, J.C. High Serum Uric Acid as a Novel Risk Factor for Type 2 Diabetes. Diabetes Care 2008, 31, 361–362. [Google Scholar] [CrossRef]
  101. Klisic, A.; Kocic, G.; Kavaric, N.; Jovanovic, M.; Stanisic, V.; Ninic, A. Xanthine oxidase and uric acid as independent predictors of albuminuria in patients with diabetes mellitus type 2. Clin. Exp. Med. 2018, 18, 283–290. [Google Scholar] [CrossRef] [PubMed]
  102. Mizuno, Y.; Yamamotoya, T.; Nakatsu, Y.; Ueda, K.; Matsunaga, Y.; Inoue, M.-K.; Sakoda, H.; Fujishiro, M.; Ono, H.; Kikuchi, T.; et al. Xanthine Oxidase Inhibitor Febuxostat Exerts an Anti-Inflammatory Action and Protects against Diabetic Nephropathy Development in KK-Ay Obese Diabetic Mice. Int. J. Mol. Sci. 2019, 20, 4680. [Google Scholar] [CrossRef] [PubMed]
  103. Wu, B.; Chen, L.; Xu, Y.; Duan, Q.; Zheng, Z.; Zheng, Z.; He, D. The Effect of Allopurinol on Renal Outcomes in Patients with Diabetic Kidney Disease: A Systematic Review and Meta-Analysis. Kidney Blood Press. Res. 2022, 47, 291–299. [Google Scholar] [CrossRef] [PubMed]
  104. Luo, Q.; Cai, Y.; Zhao, Q.; Tian, L.; Liu, Y.; Liu, W.J. Effects of allopurinol on renal function in patients with diabetes: A systematic review and meta-analysis. Ren. Fail. 2022, 44, 806–814. [Google Scholar] [CrossRef] [PubMed]
  105. Badve, S.V.; Pascoe, E.M.; Tiku, A.; Boudville, N.; Brown, F.G.; Cass, A.; Clarke, P.; Dalbeth, N.; Day, R.O.; de Zoysa, J.R.; et al. Effects of Allopurinol on the Progression of Chronic Kidney Disease. N. Engl. J. Med. 2020, 382, 2504–2513. [Google Scholar] [CrossRef] [PubMed]
  106. Doria, A.; Galecki, A.T.; Spino, C.; Pop-Busui, R.; Cherney, D.Z.; Lingvay, I.; Parsa, A.; Rossing, P.; Sigal, R.J.; Afkarian, M.; et al. Serum Urate Lowering with Allopurinol and Kidney Function in Type 1 Diabetes. New Engl. J. Med. 2020, 382, 2493–2503. [Google Scholar] [CrossRef] [PubMed]
  107. Goldberg, A.; Garcia-Arroyo, F.; Sasai, F.; Rodriguez-Iturbe, B.; Sanchez-Lozada, L.G.; Lanaspa, M.A.; Johnson, R.J. Mini Review: Reappraisal of Uric Acid in Chronic Kidney Disease. Am. J. Nephrol. 2021, 52, 837–844. [Google Scholar] [CrossRef] [PubMed]
  108. Galbusera, C.; Orth, P.; Fedida, D.; Spector, T. Superoxide radical production by allopurinol and xanthine oxidase. Biochem. Pharmacol. 2006, 71, 1747–1752. [Google Scholar] [CrossRef] [PubMed]
  109. Haberland, A.; Luther, H.; Schimke, I. Does Allopurinol Prevent Superoxide Radical Production by Xanthine Oxidase (XOD)? Agents Actions 1991, 32, 96–97. [Google Scholar] [CrossRef] [PubMed]
  110. Massey, V.; Komai, H.; Palmer, G.; Elion, G.B. On the Mechanism of Inactivation of Xanthine Oxidase by Allopurinol and Other Pyrazolo[3,4-d]pyrimidines. J. Biol. Chem. 1970, 245, 2837–2844. [Google Scholar] [CrossRef]
  111. Horiuchi, H.; Ota, M.; Nishimura, S.-I.; Kaneko, H.; Kasahara, Y.; Ohta, T.; Komoriya, K. Allopurinol induces renal toxicity by impairing pyrimidine metabolism in mice. Life Sci. 2000, 66, 2051–2070. [Google Scholar] [CrossRef] [PubMed]
  112. Takano, Y.; Hase-Aoki, K.; Horiuchi, H.; Zhao, L.; Kasahara, Y.; Kondo, S.; Becker, M.A. Selectivity of febuxostat, a novel non-purine inhibitor of xanthine oxidase/xanthine dehydrogenase. Life Sci. 2005, 76, 1835–1847. [Google Scholar] [CrossRef] [PubMed]
  113. Lee, H.-J.; Jeong, K.H.; Kim, Y.G.; Moon, J.Y.; Lee, S.H.; Ihm, C.G.; Sung, J.Y.; Lee, T.W. Febuxostat Ameliorates Diabetic Renal Injury in a Streptozotocin-Induced Diabetic Rat Model. Am. J. Nephrol. 2014, 40, 56–63. [Google Scholar] [CrossRef] [PubMed]
  114. Sircar, D.; Chatterjee, S.; Waikhom, R.; Golay, V.; Raychaudhury, A.; Chatterjee, S.; Pandey, R. Efficacy of Febuxostat for Slowing the GFR Decline in Patients With CKD and Asymptomatic Hyperuricemia: A 6-Month, Double-Blind, Randomized, Placebo-Controlled Trial. Am. J. Kidney Dis. 2015, 66, 945–950. [Google Scholar] [CrossRef] [PubMed]
  115. Kimura, K.; Hosoya, T.; Uchida, S.; Inaba, M.; Makino, H.; Ito, S.; Yamamoto, T.; Tomino, Y.; Ohno, I.; Shibagaki, Y.; et al. Febuxostat Therapy for Patients With Stage 3 CKD and Asymptomatic Hyperuricemia: A Randomized Trial. Am. J. Kidney Dis. 2018, 72, 798–810. [Google Scholar] [CrossRef] [PubMed]
  116. White, W.B.; Saag, K.G.; Becker, M.A.; Borer, J.S.; Gorelick, P.B.; Whelton, A.; Hunt, B.; Castillo, M.; Gunawardhana, L. Cardi-ovascular Safety of Febuxostat or Allopurinol in Patients with Gout. New Engl. J. Med. 2018, 378, 1200–1210. [Google Scholar] [CrossRef] [PubMed]
  117. Zheng, Y.; Sun, J. Febuxostat Improves Uric Acid Levels and Renal Function in Patients with Chronic Kidney Disease and Hyperuricemia: A Meta-Analysis. Appl. Bionics Biomech. 2022, 2022, 9704862. [Google Scholar] [CrossRef] [PubMed]
  118. Lin, T.-C.; Hung, L.Y.; Chen, Y.-C.; Lo, W.-C.; Lin, C.H.; Tam, K.-W.; Wu, M.-Y. Effects of febuxostat on renal function in patients with chronic kidney disease: A Systematic Review and Meta-Analysis. Medicine 2019, 98, e16311. [Google Scholar] [CrossRef] [PubMed]
  119. Wada, T.; Hosoya, T.; Honda, D.; Sakamoto, R.; Narita, K.; Sasaki, T.; Okui, D.; Kimura, K. Uric acid-lowering and renoprotective effects of topiroxostat, a selective xanthine oxidoreductase inhibitor, in patients with diabetic nephropathy and hyperuricemia: A randomized, double-blind, placebo-controlled, parallel-group study (UPWARD study). Clin. Exp. Nephrol. 2018, 22, 860–870. [Google Scholar] [CrossRef]
  120. Mizukoshi, T.; Kato, S.; Ando, M.; Sobajima, H.; Ohashi, N.; Naruse, T.; Saka, Y.; Shimizu, H.; Nagata, T.; Maruyama, S. Renoprotective effects of topiroxostat for Hyperuricaemic patients with overt diabetic nephropathy study (ETUDE study): A prospective, randomized, multicentre clinical trial. Nephrology 2018, 23, 1023–1030. [Google Scholar] [CrossRef]
  121. Kato, S.; Ando, M.; Mizukoshi, T.; Nagata, T.; Katsuno, T.; Kosugi, T.; Tsuboi, N.; Maruyama, S. Randomized control trial for the assessment of the anti-albuminuric effects of topiroxostat in hyperuricemic patients with diabetic nephropathy (the ETUDE study). Nagoya J. Med. Sci. 2016, 78, 135–142. [Google Scholar] [PubMed]
  122. Nakamura, T.; Murase, T.; Nampei, M.; Morimoto, N.; Ashizawa, N.; Iwanaga, T.; Sakamoto, R. Effects of topiroxostat and febuxostat on urinary albumin excretion and plasma xanthine oxidoreductase activity in db/db mice. Eur. J. Pharmacol. 2016, 780, 224–231. [Google Scholar] [CrossRef] [PubMed]
  123. Sánchez-Lozada, L.G.; Tapia, E.; Soto, V.; Ávila-Casado, C.; Franco, M.; Wessale, J.L.; Zhao, L.; Johnson, R.J. Effect of Febuxostat on the Progression of Renal Disease in 5/6 Nephrectomy Rats with and without Hyperuricemia. Nephron Physiol. 2008, 108, p69–p78. [Google Scholar] [CrossRef]
  124. Omori, H.; Kawada, N.; Inoue, K.; Ueda, Y.; Yamamoto, R.; Matsui, I.; Kaimori, J.; Takabatake, Y.; Moriyama, T.; Isaka, Y.; et al. Use of xanthine oxidase inhibitor febuxostat inhibits renal interstitial inflammation and fibrosis in unilateral ureteral obstructive nephropathy. Clin. Exp. Nephrol. 2012, 16, 549–556. [Google Scholar] [CrossRef] [PubMed]
  125. Miura, T.; Sakuyama, A.; Xu, L.; Qiu, J.; Namai-Takahashi, A.; Ogawa, Y.; Kohzuki, M.; Ito, O. Febuxostat ameliorates high salt intake-induced hypertension and renal damage in Dahl salt-sensitive rats. J. Hypertens. 2021, 40, 327–337. [Google Scholar] [CrossRef] [PubMed]
  126. Inoue, M.-K.; Yamamotoya, T.; Nakatsu, Y.; Ueda, K.; Inoue, Y.; Matsunaga, Y.; Sakoda, H.; Fujishiro, M.; Ono, H.; Morii, K.; et al. The Xanthine Oxidase Inhibitor Febuxostat Suppresses the Progression of IgA Nephropathy, Possibly via Its Anti-Inflammatory and Anti-Fibrotic Effects in the gddY Mouse Model. Int. J. Mol. Sci. 2018, 19, 3967. [Google Scholar] [CrossRef] [PubMed]
  127. He, L.; Fan, Y.; Xiao, W.; Chen, T.; Wen, J.; Dong, Y.; Wang, Y.; Li, S.; Xue, R.; Zheng, L.; et al. Febuxostat attenuates ER stress mediated kidney injury in a rat model of hyperuricemic nephropathy. Oncotarget 2017, 8, 111295–111308. [Google Scholar] [CrossRef]
  128. Lim, A.K.H.; Tesch, G.H. Inflammation in Diabetic Nephropathy. Mediat. Inflamm. 2013, 2012, 146154. [Google Scholar] [CrossRef] [PubMed]
  129. Liu, P.; Chen, Y.; Wang, B.; Zhang, F.; Wang, D.; Wang, Y. Allopurinol treatment improves renal function in patients with type 2 diabetes and asymptomatic hyperuricemia: 3-year randomized parallel-controlled study. Clin. Endocrinol. 2015, 83, 475–482. [Google Scholar] [CrossRef]
  130. Tanaka, K.; Nakayama, M.; Kanno, M.; Kimura, H.; Watanabe, K.; Tani, Y.; Hayashi, Y.; Asahi, K.; Terawaki, H.; Watanabe, T. Renoprotective effects of febuxostat in hyperuricemic patients with chronic kidney disease: A parallel-group, randomized, controlled trial. Clin. Exp. Nephrol. 2015, 19, 1044–1053. [Google Scholar] [CrossRef]
  131. Hosoya, T.; Ohno, I.; Nomura, S.; Hisatome, I.; Uchida, S.; Fujimori, S.; Yamamoto, T.; Hara, S. Effects of topiroxostat on the serum urate levels and urinary albumin excretion in hyperuricemic stage 3 chronic kidney disease patients with or without gout. Clin. Exp. Nephrol. 2014, 18, 876–884. [Google Scholar] [CrossRef] [PubMed]
  132. Nata, N.; Ninwisut, N.; Inkong, P.; Supasyndh, O.; Satirapoj, B. Effects of febuxostat on markers of endothelial dysfunction and renal progression in patients with chronic kidney disease. Sci. Rep. 2023, 13, 13494. [Google Scholar] [CrossRef] [PubMed]
  133. Ejaz, A.A.; Antenor, J.A.; Kumar, V.; Roncal, C.; Garcia, G.E.; Andres-Hernando, A.; Lanaspa, M.A.; Johnson, R.J. Uric Acid: A Friend in the Past, a Foe in the Present. Integr. Med. Nephrol. Androl. 2022, 9, 8. [Google Scholar] [CrossRef]
  134. Yang, J.; Kamide, K.; Kokubo, Y.; Takiuchi, S.; Horio, T.; Matayoshi, T.; Yasuda, H.; Miwa, Y.; Yoshii, M.; Yoshihara, F.; et al. Assiciations of Hypertension and Its Complications with Variations in the Xanthine Dehydrogenase Gene. Hypertens. Res. 2008, 31, 931–940. [Google Scholar] [CrossRef] [PubMed]
  135. Kocyigit, I.; Yilmaz, M.I.; Orscelik, O.; Sipahioglu, M.H.; Unal, A.; Eroglu, E.; Kalay, N.; Tokgoz, B.; Axelsson, J.; Oymak, O. Serum Uric Acid Levels and Endothelial Dysfunction in Patients with Autosomal Dominant Polycystic Kidney Disease. Nephron Clin. Pract. 2013, 123, 157–164. [Google Scholar] [CrossRef] [PubMed]
  136. Guay-Woodford, L.M.; Desmond, R.A. Autosomal Recessive Polycystic Kidney Disease: The Clinical Experience in North America. Pediatrics 2003, 111, 1072–1080. [Google Scholar] [CrossRef]
  137. Torra, R.; Pérez-Gómez, M.V.; Furlano, M. Autosomal dominant polycystic kidney disease: Possibly the least silent cause of chronic kidney disease. Clin. Kidney J. 2021, 14, 2281–2284. [Google Scholar] [CrossRef]
  138. Menon, V.; Rudym, D.; Chandra, P.; Miskulin, D.; Perrone, R.; Sarnak, M. Inflammation, Oxidative Stress, and Insulin Resistance in Polycystic Kidney Disease. Clin. J. Am. Soc. Nephrol. 2011, 6, 7–13. [Google Scholar] [CrossRef]
  139. Vareesangthip, K.; Tong, P.; Wilkinson, R.; Thomas, T.H. Insulin resistance in adult polycystic kidney disease. Kidney Int. 1997, 52, 503–508. [Google Scholar] [CrossRef]
  140. Kaehny, W.D.; Tangel, D.J.; Johnson, A.M.; Kimberling, W.J.; Schrier, R.W.; Gabow, P.A. Uric acid handling in autosomal dominant polycystic kidney disease with normal filtration rates. Am. J. Med. 1990, 89, 49–52. [Google Scholar] [CrossRef]
  141. Kim, H.; Koh, J.; Park, S.K.; Oh, K.H.; Kim, Y.H.; Kim, Y.; Ahn, C.; Oh, Y.K. Baseline characteristics of the autosomal-dominant polycystic kidney disease sub-cohort of the KoreaN cohort study for outcomes in patients with chronic kidney disease. Nephrology 2019, 24, 422–429. [Google Scholar] [CrossRef] [PubMed]
  142. Wu, B.; Roseland, J.M.; Haytowitz, D.B.; Pehrsson, P.R.; Ershow, A.G. Availability and quality of published data on the purine content of foods, alcoholic beverages, and dietary supplements. J. Food Compos. Anal. 2019, 84, 103281. [Google Scholar] [CrossRef]
  143. Zhang, P.; Sun, H.; Cheng, X.; Li, Y.; Zhao, Y.; Mei, W.; Wei, X.; Zhou, H.; Du, Y.; Zeng, C. Dietary intake of fructose increases purine de novo synthesis: A crucial mechanism for hyperuricemia. Front. Nutr. 2022, 9, 1045805. [Google Scholar] [CrossRef] [PubMed]
  144. Vedder, D.; Walrabenstein, W.; Heslinga, M.; de Vries, R.; Nurmohamed, M.; van Schaardenburg, D.; Gerritsen, M. Dietary Interventions for Gout and Effect on Cardiovascular Risk Factors: A Systematic Review. Nutrients 2019, 11, 2955. [Google Scholar] [CrossRef] [PubMed]
  145. Aihemaitijiang, S.; Zhang, Y.; Zhang, L.; Yang, J.; Ye, C.; Halimulati, M.; Zhang, W.; Zhang, Z. The Association between Purine-Rich Food Intake and Hyperuricemia: A Cross-Sectional Study in Chinese Adult Residents. Nutrients 2020, 12, 3835. [Google Scholar] [CrossRef] [PubMed]
  146. Pang, B.; McFaline, J.L.; Burgis, N.E.; Dong, M.; Taghizadeh, K.; Sullivan, M.R.; Elmquist, C.E.; Cunningham, R.P.; Dedon, P.C. Defects in purine nucleotide metabolism lead to substantial incorporation of xanthine and hypoxanthine into DNA and RNA. Proc. Natl. Acad. Sci. USA 2012, 109, 2319–2324. [Google Scholar] [CrossRef] [PubMed]
  147. Pareek, V.; Pedley, A.M.; Benkovic, S.J. Human de novo purine biosynthesis. Crit. Rev. Biochem. Mol. Biol. 2021, 56, 1–16. [Google Scholar] [CrossRef] [PubMed]
  148. Major, T.J.; Topless, R.K.; Dalbeth, N.; Merriman, T.R. Evaluation of the diet wide contribution to serum urate levels: Meta-analysis of population based cohorts. BMJ 2018, 363, k3951. [Google Scholar] [CrossRef] [PubMed]
  149. Rai, S.K.; Fung, T.T.; Lu, N.; Keller, S.F.; Curhan, G.C.; Choi, H.K. The Dietary Approaches to Stop Hypertension (DASH) diet, Western diet, and risk of gout in men: Prospective cohort study. BMJ 2017, 357, j1794. [Google Scholar] [CrossRef]
  150. Clemente-Suárez, V.J.; Beltrán-Velasco, A.I.; Redondo-Flórez, L.; Martín-Rodríguez, A.; Tornero-Aguilera, J.F. Global Impacts of Western Diet and Its Effects on Metabolism and Health: A Narrative Review. Nutrients 2023, 15, 2749. [Google Scholar] [CrossRef]
  151. Kopp, W. How Western Diet And Lifestyle Drive The Pandemic Of Obesity And Civilization Diseases. Diabetes Metab. Syndr. Obes. Targets Ther. 2019, 12, 2221–2236. [Google Scholar] [CrossRef]
  152. Argueta, D.A.; DiPatrizio, N.V. Peripheral endocannabinoid signaling controls hyperphagia in western diet-induced obesity. Physiol. Behav. 2017, 171, 32–39. [Google Scholar] [CrossRef]
  153. Stevenson, R.J.; Francis, H.M.; Attuquayefio, T.; Gupta, D.; Yeomans, M.R.; Oaten, M.J.; Davidson, T. Hippocampal-dependent appetitive control is impaired by experimental exposure to a Western-style diet. R. Soc. Open Sci. 2020, 7, 191338. [Google Scholar] [CrossRef]
  154. Webb, A.; Bond, R.; McLean, P.; Uppal, R.; Benjamin, N.; Ahluwalia, A. Reduction of nitrite to nitric oxide during ischemia protects against myocardial ischemia–reperfusion damage. Proc. Natl. Acad. Sci. USA 2004, 101, 13683–13688. [Google Scholar] [CrossRef]
  155. Godber, B.L.J.; Doel, J.J.; Sapkota, G.P.; Blake, D.R.; Stevens, C.R.; Eisenthal, R.; Harrison, R. Reduction of Nitrite to Nitric Oxide Catalyzed by Xanthine Oxidoreductase. J. Biol. Chem. 2000, 275, 7757–7763. [Google Scholar] [CrossRef]
  156. Peleli, M.; Zollbrecht, C.; Montenegro, M.F.; Hezel, M.; Zhong, J.; Persson, E.G.; Holmdahl, R.; Weitzberg, E.; Lundberg, J.O.; Carlström, M. Enhanced XOR activity in eNOS-deficient mice: Effects on the Nitrate-Nitrite-NO Pathway and ROS Homeostasis. Free Radic. Biol. Med. 2016, 99, 472–484. [Google Scholar] [CrossRef] [PubMed]
  157. Danaei, G.; Singh, G.M.; Paciorek, C.J.; Lin, J.K.; Cowan, M.J.; Finucane, M.M.; Farzadfar, F.; Stevens, G.A.; Riley, L.M.; Lu, Y.; et al. The Global Cardiovascular Risk Transition: Associations of Four Metabolic Risk Factors with National Income, Urbanization, and Western Diet in 1980 and 2008. Circulation 2013, 127, 1493–1502. [Google Scholar] [CrossRef]
  158. Tikellis, C.; Thomas, M.C.; Harcourt, B.E.; Coughlan, M.T.; Pete, J.; Bialkowski, K.; Tan, A.; Bierhaus, A.; Cooper, M.E.; Forbes, J.M.; et al. Cardiac inflammation associated with a Western diet is mediated via activation of RAGE by AGEs. Am. J. Physiol. Endocrinol. Metab. 2008, 295, E323–E330. [Google Scholar] [CrossRef] [PubMed]
  159. Ballal, K.; Wilson, C.R.; Harmancey, R.; Taegtmeyer, H. Obesogenic high fat western diet induces oxidative stress and apoptosis in rat heart. Mol. Cell. Biochem. 2010, 344, 221–230. [Google Scholar] [CrossRef]
  160. Aroor, A.R.; Jia, G.; Habibi, J.; Sun, Z.; Ramirez-Perez, F.I.; Brady, B.; Chen, D.; Martinez-Lemus, L.A.; Manrique, C.; Nistala, R.; et al. Uric acid promotes vascular stiffness, maladaptive inflammatory responses and proteinuria in western diet fed mice. Metabolism 2017, 74, 32–40. [Google Scholar] [CrossRef]
  161. Akki, A.; Seymour, A.-M.L. Western diet impairs metabolic remodelling and contractile efficiency in cardiac hypertrophy. Cardiovasc. Res. 2009, 81, 610–617. [Google Scholar] [CrossRef] [PubMed]
  162. Schröder, K.; Vecchione, C.; Jung, O.; Schreiber, J.G.; Shiri-Sverdlov, R.; van Gorp, P.J.; Busse, R.; Brandes, R.P. Xanthine oxidase inhibitor tungsten prevents the development of atherosclerosis in ApoE knockout mice fed a Western-type diet. Free. Radic. Biol. Med. 2006, 41, 1353–1360. [Google Scholar] [CrossRef] [PubMed]
  163. Salehi-Sahlabadi, A.; Sadat, S.; Beigrezaei, S.; Pourmasomi, M.; Feizi, A.; Ghiasvand, R.; Hadi, A.; Clark, C.C.T.; Miraghajani, M. Dietary patterns and risk of non-alcoholic fatty liver disease. BMC Gastroenterol. 2021, 21, 41. [Google Scholar] [CrossRef] [PubMed]
  164. Eslam, M.; Sanyal, A.J.; George, J.; on behalf of the International Consensus Panel. MAFLD: A Consensus-Driven Proposed Nomenclature for Metabolic Associated Fatty Liver Disease. Gastroenterology 2020, 158, 1999–2014.e1. [Google Scholar] [CrossRef] [PubMed]
  165. Yagi, C.; Kusunoki, Y.; Tsunoda, T.; Murase, T.; Nakamura, T.; Osugi, K.; Ohigashi, M.; Morimoto, A.; Miyoshi, A.; Kakutani-Hatayama, M.; et al. Xanthine oxidoreductase activity is correlated with hepatic steatosis. Sci. Rep. 2022, 12, 12282. [Google Scholar] [CrossRef]
  166. Demaria, T.M.; Crepaldi, L.D.; Costa-Bartuli, E.; Branco, J.R.; Zancan, P.; Sola-Penna, M. Once a week consumption of Western diet over twelve weeks promotes sustained insulin resistance and non-alcoholic fat liver disease in C57BL/6 J mice. Sci. Rep. 2023, 13, 3058. [Google Scholar] [CrossRef]
  167. Kawachi, Y.; Fujishima, Y.; Nishizawa, H.; Nakamura, T.; Akari, S.; Murase, T.; Saito, T.; Miyazaki, Y.; Nagao, H.; Fukuda, S.; et al. Increased plasma XOR activity induced by NAFLD/NASH and its possible involvement in vascular neointimal proliferation. J. Clin. Investig. 2021, 6, e144762. [Google Scholar] [CrossRef]
  168. Nasiri-Ansari, N.; Androutsakos, T.; Flessa, C.-M.; Kyrou, I.; Siasos, G.; Randeva, H.S.; Kassi, E.; Papavassiliou, A.G. Endothelial Cell Dysfunction and Nonalcoholic Fatty Liver Disease (NAFLD): A Concise Review. Cells 2022, 11, 2511. [Google Scholar] [CrossRef]
  169. Johnson, R.J.; Perez-Pozo, S.E.; Lillo, J.L.; Grases, F.; Schold, J.D.; Kuwabara, M.; Sato, Y.; Hernando, A.A.; Garcia, G.; Jensen, T.; et al. Fructose increases risk for kidney stones: Potential role in metabolic syndrome and heat stress. BMC Nephrol. 2018, 19, 315. [Google Scholar] [CrossRef]
  170. Li, X.; Bhattacharya, D.; Yuan, Y.; Wei, C.; Zhong, F.; Ding, F.; D’agati, V.D.; Lee, K.; Friedman, S.L.; He, J.C. Chronic kidney disease in a murine model of non-alcoholic steatohepatitis (NASH). Kidney Int. 2024, 105, 540–561. [Google Scholar] [CrossRef]
  171. Nakagawa, T.; Hu, H.; Zharikov, S.; Tuttle, K.R.; Short, R.A.; Glushakova, O.; Ouyang, X.; Feig, D.I.; Block, E.R.; Herrera-Acosta, J.; et al. A causal role for uric acid in fructose-induced metabolic syndrome. Am. J. Physiol. Physiol. 2006, 290, F625–F631. [Google Scholar] [CrossRef] [PubMed]
  172. Nakagawa, T.; Johnson, R.J.; Andres-Hernando, A.; Roncal-Jimenez, C.; Sanchez-Lozada, L.G.; Tolan, D.R.; Lanaspa, M.A. Fructose Production and Metabolism in the Kidney. J. Am. Soc. Nephrol. 2020, 31, 898–906. [Google Scholar] [CrossRef] [PubMed]
  173. Nakayama, T.; Kosugi, T.; Gersch, M.; Connor, T.; Sanchez-Lozada, L.G.; Lanaspa, M.A.; Roncal, C.; Perez-Pozo, S.E.; Johnson, R.J.; Nakagawa, T.; et al. Dietary fructose causes tubulointerstitial injury in the normal rat kidney. Am. J. Physiol. Renal Physiol. 2010, 298, F712–F720. [Google Scholar] [CrossRef] [PubMed]
  174. Nakagawa, T.; Kang, D.-H. Fructose in the kidney: From physiology to pathology. Kidney Res. Clin. Pract. 2021, 40, 527–541. [Google Scholar] [CrossRef]
  175. Cirillo, P.; Gersch, M.S.; Mu, W.; Scherer, P.M.; Kim, K.M.; Gesualdo, L.; Henderson, G.N.; Johnson, R.J.; Sautin, Y.Y. Ketohexokinase-Dependent Metabolism of Fructose Induces Proinflammatory Mediators in Proximal Tubular Cells. J. Am. Soc. Nephrol. 2009, 20, 545–553. [Google Scholar] [CrossRef] [PubMed]
  176. Mohamad, H.E.; Abdelhady, M.A.; Aal, S.M.A.; Elrashidy, R.A. Dulaglutide mitigates high dietary fructose-induced renal fibrosis in rats through suppressing epithelial-mesenchymal transition mediated by GSK-3β/TGF-β1/Smad3 signaling pathways. Life Sci. 2022, 309, 120999. [Google Scholar] [CrossRef] [PubMed]
  177. Peng, Z.-Y.; Yang, C.-T.; Lin, W.-H.; Yao, W.-Y.; Ou, H.-T.; Kuo, S. Chronic kidney outcomes associated with GLP-1 receptor agonists versus long-acting insulins among type 2 diabetes patients requiring intensive glycemic control: A nationwide cohort study. Cardiovasc. Diabetol. 2023, 22, 272. [Google Scholar] [CrossRef] [PubMed]
  178. Odermatt, A. The Western-style diet: A major risk factor for impaired kidney function and chronic kidney disease. Am. J. Physiol. Renal Physiol. 2011, 301, F919–F931. [Google Scholar] [CrossRef] [PubMed]
  179. Zhang, M.; Chang, H.; Gao, Y.; Wang, X.; Xu, W.; Liu, D.; Li, G.; Huang, G. Major Dietary Patterns and Risk of Asymptomatic Hyperuricemia in Chinese Adults. J. Nutr. Sci. Vitaminol. 2012, 58, 339–345. [Google Scholar] [CrossRef]
  180. Suzuki, H.; DeLano, F.A.; Parks, D.A.; Jamshidi, N.; Granger, D.N.; Ishii, H.; Suematsu, M.; Zweifach, B.W.; Schmid-Schönbein, G.W. Xanthine oxidase activity associated with arterial blood pressure in spontaneously hypertensive rats. Proc. Natl. Acad. Sci. USA 1998, 95, 4754–4759. [Google Scholar] [CrossRef]
  181. Swei, A.; Lacy, F.; Delano, F.A.; Parks, D.A.; Schmid-Schönbein, G.W. A Mechanism of Oxygen Free Radical Production in the Dahl Hypertensive Rat. Microcirculation 1999, 6, 179–187. [Google Scholar] [CrossRef] [PubMed]
  182. Wesseling, C.; Aragón, A.; González, M.; Weiss, I.; Glaser, J.; Rivard, C.J.; Roncal-Jiménez, C.; Correa-Rotter, R.; Johnson, R.J. Heat stress, hydration and uric acid: A cross-sectional study in workers of three occupations in a hotspot of Mesoamerican nephropathy in Nicaragua. BMJ Open 2016, 6, e011034. [Google Scholar] [CrossRef]
  183. Ichida, K.; Amaya, Y.; Kamatani, N.; Nishino, T.; Hosoya, T.; Sakai, O. Identification of two mutations in human xanthine dehydrogenase gene responsible for classical type I xanthinuria. J. Clin. Investig. 1997, 99, 2391–2397. [Google Scholar] [CrossRef] [PubMed]
  184. Liu, H.; Zhu, H.; Shi, W.; Lin, Y.; Ma, G.; Tao, G.; Gong, W.; Zhao, Q.; Du, M.; Wang, M.; et al. Genetic variants in XDH are associated with prognosis for gastric cancer in a Chinese population. Gene 2018, 663, 196–202. [Google Scholar] [CrossRef] [PubMed]
  185. Steunou, A.-S.; Babot, M.; Durand, A.; Bourbon, M.-L.; Liotenberg, S.; Miotello, G.; Armengaud, J.; Ouchane, S. Discriminating Susceptibility of Xanthine Oxidoreductase Family to Metals. Microbiol. Spectr. 2023, 11, e0481422. [Google Scholar] [CrossRef] [PubMed]
  186. Roncal-Jimenez, C.A.; Sato, Y.; Milagres, T.; Hernando, A.A.; García, G.; Bjornstad, P.; Dawson, J.B.; Sorensen, C.; Newman, L.; Krisher, L.; et al. Experimental heat stress nephropathy and liver injury are improved by allopurinol. Am. J. Physiol. Renal Physiol. 2018, 315, F726–F733. [Google Scholar] [CrossRef] [PubMed]
  187. Crane, J.K.; Naeher, T.M.; Broome, J.E.; Boedeker, E.C. Role of Host Xanthine Oxidase in Infection Due to Enteropathogenic and Shiga-Toxigenic Escherichia coli. Infect. Immun. 2013, 81, 1129–1139. [Google Scholar] [CrossRef] [PubMed]
  188. Xu, J.; Zhu, X.; Hui, R.; Xing, Y.; Wang, J.; Shi, S.; Zhang, Y.; Zhu, L. Associations of metal exposure with hyperuricemia and gout in general adults. Front. Endocrinol. 2022, 13, 1052784. [Google Scholar] [CrossRef]
  189. Madigan, M.C.; McEnaney, R.M.; Shukla, A.J.; Hong, G.; Kelley, E.E.; Tarpey, M.M.; Gladwin, M.; Zuckerbraun, B.S.; Tzeng, E. Xanthine Oxidoreductase Function Contributes to Normal Wound Healing. Mol. Med. 2015, 21, 313–322. [Google Scholar] [CrossRef]
  190. Sorensen, C.; Garcia-Trabanino, R. A New Era of Climate Medicine—Addressing Heat-Triggered Renal Disease. New Engl. J. Med. 2019, 381, 693–696. [Google Scholar] [CrossRef]
  191. Hansson, E.; Glaser, J.; Jakobsson, K.; Weiss, I.; Wesseling, C.; Lucas, R.A.I.; Wei, J.L.K.; Ekström, U.; Wijkström, J.; Bodin, T.; et al. Pathophysiological Mechanisms by which Heat Stress Potentially Induces Kidney Inflammation and Chronic Kidney Disease in Sugarcane Workers. Nutrients 2020, 12, 1639. [Google Scholar] [CrossRef] [PubMed]
  192. Trabanino, R.G.; Aguilar, R.; Silva, C.R.; Mercado, M.O.; Merino, R.L. Nefropatía terminal en pacientes de un hospital de referencia en El Salvador. Rev. Panam. De Salud Publica-Pan Am. J. Public Health 2002, 12, 202–206. [Google Scholar] [CrossRef]
  193. John, O.; Gummudi, B.; Jha, A.; Gopalakrishnan, N.; Kalra, O.P.; Kaur, P.; Kher, V.; Kumar, V.; Machiraju, R.S.; Osborne, N.; et al. Chronic Kidney Disease of Unknown Etiology in India: What Do We Know and Where We Need to Go. Kidney Int. Rep. 2021, 6, 2743–2751. [Google Scholar] [CrossRef]
  194. Mani, M.K. Chronic renal failure in India. Nephrol. Dial. Transplant. 1993, 8, 684–689. [Google Scholar] [CrossRef] [PubMed]
  195. Rajapakse, S.; Shivanthan, M.C.; Selvarajah, M. Chronic kidney disease of unknown etiology in Sri Lanka. Int. J. Occup. Environ. Health 2016, 22, 259–264. [Google Scholar] [CrossRef]
  196. Roncal-Jimenez, C.; García-Trabanino, R.; Barregard, L.; Lanaspa, M.A.; Wesseling, C.; Harra, T.; Aragón, A.; Grases, F.; Jarquin, E.R.; González, M.A.; et al. Heat Stress Nephropathy From Exercise-Induced Uric Acid Crystalluria: A Perspective on Mesoamerican Nephropathy. Am. J. Kidney Dis. 2016, 67, 20–30. [Google Scholar] [CrossRef]
  197. Laws, R.L.; Brooks, D.R.; Amador, J.J.; Weiner, D.E.; Kaufman, J.S.; Ramírez-Rubio, O.; Riefkohl, A.; Scammell, M.K.; López-Pilarte, D.; Sánchez, J.M.; et al. Changes in kidney function among Nicaraguan sugarcane workers. Int. J. Occup. Environ. Health 2015, 21, 241–250. [Google Scholar] [CrossRef]
  198. Wijkström, J.; Leiva, R.; Elinder, C.-G.; Leiva, S.; Trujillo, Z.; Trujillo, L.; Söderberg, M.; Hultenby, K.; Wernerson, A. Clinical and Pathological Characterization of Mesoamerican Nephropathy: A New Kidney Disease in Central America. Am. J. Kidney Dis. 2013, 62, 908–918. [Google Scholar] [CrossRef]
  199. Ryu, E.-S.; Kim, M.J.; Shin, H.-S.; Jang, Y.-H.; Choi, H.S.; Jo, I.; Johnson, R.J.; Kang, D.-H. Uric acid-induced phenotypic transition of renal tubular cells as a novel mechanism of chronic kidney disease. Am. J. Physiol. Ren. Physiol. 2013, 304, F471–F480. [Google Scholar] [CrossRef]
  200. Sorensen, C.J.; Butler-Dawson, J.; Dally, M.; Krisher, L.; Griffin, B.R.; Johnson, R.J.; Lemery, J.; Asensio, C.; Tenney, L.; Newman, L.S. Risk Factors and Mechanisms Underlying Cross-Shift Decline in Kidney Function in Guatemalan Sugarcane Workers. J. Occup. Environ. Med. 2019, 61, 239–250. [Google Scholar] [CrossRef]
  201. Wesseling, C.; Glaser, J.; Rodríguez-Guzmán, J.; Weiss, I.; Lucas, R.; Peraza, S.; da Silva, A.S.; Hansson, E.; Johnson, R.J.; Hogstedt, C.; et al. Chronic kidney disease of non-traditional origin in Mesoamerica: A disease primarily driven by occupational heat stress. Rev. Panam. Salud Publica—Pan Am. J. Public Health 2020, 44, e15. [Google Scholar] [CrossRef] [PubMed]
  202. Wei, X.-Y.; Jia, P.-P.; Hu, H.; Liu, L.; Li, T.-Y.; Li, Y.-Z.; Pei, D.-S. Multi-omics reveal mechanisms underlying chronic kidney disease of unknown etiology (CKDu) pathogenesis using zebrafish. Environ. Pollut. 2023, 337, 122524. [Google Scholar] [CrossRef]
  203. Schaeffer, J.W.; Adgate, J.L.; Reynolds, S.J.; Butler-Dawson, J.; Krisher, L.; Dally, M.; Johnson, R.J.; James, K.A.; Jaramillo, D.; Newman, L.S. A Pilot Study to Assess Inhalation Exposures among Sugarcane Workers in Guatemala: Implications for Chronic Kidney Disease of Unknown Origin. Int. J. Environ. Res. Public Health 2020, 17, 5708. [Google Scholar] [CrossRef]
  204. Valcke, M.; Levasseur, M.-E.; da Silva, A.S.; Wesseling, C. Pesticide exposures and chronic kidney disease of unknown etiology: An epidemiologic review. Environ. Health 2017, 16, 49. [Google Scholar] [CrossRef]
  205. Butler-Dawson, J.; James, K.A.; Krisher, L.; Jaramillo, D.; Dally, M.; Neumann, N.; Pilloni, D.; Cruz, A.; Asensio, C.; Johnson, R.J.; et al. Environmental metal exposures and kidney function of Guatemalan sugarcane workers. J. Expo. Sci. Environ. Epidemiology 2021, 32, 461–471. [Google Scholar] [CrossRef]
  206. Rodrigues, P.; Furriol, J.; Bermejo, B.; Chaves, F.J.; Lluch, A.; Eroles, P. Identification of Candidate Polymorphisms on Stress Oxidative and DNA Damage Repair Genes Related with Clinical Outcome in Breast Cancer Patients. Int. J. Mol. Sci. 2012, 13, 16500–16513. [Google Scholar] [CrossRef] [PubMed]
  207. Rodrigues, P.; de Marco, G.; Furriol, J.; Mansego, M.L.; Pineda-Alonso, M.; Gonzalez-Neira, A.; Martin-Escudero, J.C.; Benitez, J.; Lluch, A.; Chaves, F.J.; et al. Oxidative stress in susceptibility to breast cancer: Study in Spanish population. BMC Cancer 2014, 14, 861. [Google Scholar] [CrossRef] [PubMed]
  208. Xu, P.; LaVallee, P.; Hoidal, J.R. Repressed Expression of the Human Xanthine Oxidoreductase Gene. E-Box and TATA-like Elements Restrict Ground State Transcriptional Activity. J. Biol. Chem. 2000, 275, 5918–5926. [Google Scholar] [CrossRef] [PubMed]
  209. Kudo, M.; Sasaki, T.; Ishikawa, M.; Hirasawa, N.; Hiratsuka, M. Functional Characterization of Genetic Polymorphisms Identified in the Promoter Region of the Xanthine Oxidase Gene. Drug Metab. Pharmacokinet. 2010, 25, 599–604. [Google Scholar] [CrossRef]
  210. Boban, M.; Kocic, G.; Radenkovic, S.; Pavlovic, R.; Cvetkovic, T.; Deljanin-Ilic, M.; Ilic, S.; Bobana, M.D.; Djindjic, B.; Stojanovic, D.; et al. Circulating purine compounds, uric acid, and xanthine oxidase/dehydrogenase relationship in essential hypertension and end stage renal disease. Ren. Fail. 2014, 36, 613–618. [Google Scholar] [CrossRef]
  211. Fujishiro, M.; Ishihara, H.; Ogawa, K.; Murase, T.; Nakamura, T.; Watanabe, K.; Sakoda, H.; Ono, H.; Yamamotoya, T.; Nakatsu, Y.; et al. Impact of Plasma Xanthine Oxidoreductase Activity on the Mechanisms of Distal Symmetric Polyneuropathy Development in Patients with Type 2 Diabetes. Biomedicines 2021, 9, 1052. [Google Scholar] [CrossRef]
  212. Okuyama, T.; Shirakawa, J.; Nakamura, T.; Murase, T.; Miyashita, D.; Inoue, R.; Kyohara, M.; Togashi, Y.; Terauchi, Y. Association of the plasma xanthine oxidoreductase activity with the metabolic parameters and vascular complications in patients with type 2 diabetes. Sci. Rep. 2021, 11, 3768. [Google Scholar] [CrossRef] [PubMed]
  213. Zhu, D.-D.; Wang, Y.-Z.; Zou, C.; She, X.-P.; Zheng, Z. The role of uric acid in the pathogenesis of diabetic retinopathy based on Notch pathway. Biochem. Biophys. Res. Commun. 2018, 503, 921–929. [Google Scholar] [CrossRef]
  214. Stamp, L.K.; Chapman, P.T. Allopurinol hypersensitivity: Pathogenesis and prevention. Best Pract. Res. Clin. Rheumatol. 2020, 34, 101501. [Google Scholar] [CrossRef] [PubMed]
  215. FDA. FDA Adds Boxed Warning for Increased Risk of Death with Gout Medicine Uloric (Febuxostat). Available online: https://www.fda.gov/drugs/drug-safety-and-availability/fda-adds-boxed-warning-increased-risk-death-gout-medicine-uloric-febuxostat (accessed on 2 June 2024).
  216. Rullo, R.; Cerchia, C.; Nasso, R.; Romanelli, V.; De Vendittis, E.; Masullo, M.; Lavecchia, A. Novel Reversible Inhibitors of Xanthine Oxidase Targeting the Active Site of the Enzyme. Antioxidants 2023, 12, 825. [Google Scholar] [CrossRef]
  217. Zhu, X.; Yang, C.; Zhang, L.; Li, J. Identification of novel dual inhibitors targeting XOR and URAT1 via multiple virtual screening methods. J. Mol. Struct. 2022, 1256, 132567. [Google Scholar] [CrossRef]
  218. Singh, A.; Singh, K.; Sharma, A.; Kaur, K.; Chadha, R.; Bedi, P.M.S. Past, present and future of xanthine oxidase inhibitors: Design strategies, structural and pharmacological insights, patents and clinical trials. RSC Med. Chem. 2023, 14, 2155–2191. [Google Scholar] [CrossRef]
  219. Sundy, J.S.; Baraf, H.S.B.; Yood, R.A.; Edwards, N.L.; Gutierrez-Urena, S.R.; Treadwell, E.L.; Vázquez-Mellado, J.; White, W.B.; Lipsky, P.E.; Horowitz, Z.; et al. Efficacy and Tolerability of Pegloticase for the Treatment of Chronic Gout in Patients Refractory to Conventional Treatment: Two Randomized Controlled Trials. JAMA 2011, 306, 711–720. [Google Scholar] [CrossRef]
  220. Li, Z.; Liu, Q.; Mao, H.; Li, Z.; Dong, X.; Liu, Y.; Lin, J.; Chen, W.; Wang, H.; Johnson, R.J.; et al. Gender Difference in the Association of Hyperuricemia with Chronic Kidney Disease in Southern China. Kidney Blood Press. Res. 2012, 36, 98–106. [Google Scholar] [CrossRef]
  221. Bolognesi, A.; Bortolotti, M.; Battelli, M.G.; Polito, L. Gender Influence on XOR Activities and Related Pathologies: A Narrative Review. Antioxidants 2024, 13, 211. [Google Scholar] [CrossRef]
  222. Furuhashi, M.; Higashiura, Y.; Koyama, M.; Tanaka, M.; Murase, T.; Nakamura, T.; Akari, S.; Sakai, A.; Mori, K.; Ohnishi, H.; et al. Independent association of plasma xanthine oxidoreductase activity with hypertension in nondiabetic subjects not using medication. Hypertens. Res. 2021, 44, 1213–1220. [Google Scholar] [CrossRef] [PubMed]
  223. Hasan, M.; Fariha, K.A.; Barman, Z.; Mou, A.D.; Miah, R.; Habib, A.; Tuba, H.R.; Ali, N. Assessment of the relationship between serum xanthine oxidase levels and type 2 diabetes: A cross-sectional study. Sci. Rep. 2022, 12, 20816. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Terminal Steps of Purine Catabolism Mediated by XOR. Purines are predominantly metabolized to hypoxanthine, which is subsequently oxidized to xanthine and further to UA. Inhibitors at various steps are indicated (Blue), and purine mimetics have off-target effects (Blue, thin print). The pseudogenized human UOX previously produced allantoin (Grey). Uricase therapy (Green) can promote this pathway in humans. XOR, xanthine oxidoreductase; XDH, xanthine dehydrogenase; XO, xanthine oxidase; UA, uric acid; ASL, adenine succinate lyase; ASS, adenine succinate synthase; NAD(H), nicotinamide adenine dinucleotide; PPi, inorganic pyrophosphate; UOX, urate oxidase.
Figure 1. Terminal Steps of Purine Catabolism Mediated by XOR. Purines are predominantly metabolized to hypoxanthine, which is subsequently oxidized to xanthine and further to UA. Inhibitors at various steps are indicated (Blue), and purine mimetics have off-target effects (Blue, thin print). The pseudogenized human UOX previously produced allantoin (Grey). Uricase therapy (Green) can promote this pathway in humans. XOR, xanthine oxidoreductase; XDH, xanthine dehydrogenase; XO, xanthine oxidase; UA, uric acid; ASL, adenine succinate lyase; ASS, adenine succinate synthase; NAD(H), nicotinamide adenine dinucleotide; PPi, inorganic pyrophosphate; UOX, urate oxidase.
Antioxidants 13 00712 g001
Figure 2. XOR Impacts Endothelial Structure & Function. XOR is released by multiple sources, predominantly the liver. Upon release, XOR enters the vasculature and binds to the extracellular GAG-rich glycocalyx produced by endothelial cells. Inset: XOR can contribute to the breakdown of the glycocalyx initiate endothelial activation and promote intracellular ROS production. XOR can also be endocytosed following glycocalyx binding, which may impact endothelial cell function. Additionally, XOR functions to uncouple eNOS, inhibiting NO production, and instigating ONOO- formation. eNOS, endothelial nitric oxide synthase; GAG, glycosaminoglycan; ROS, reactive oxygen species; XOR, xanthine oxidoreductase; ONOO-, peroxynitrite.
Figure 2. XOR Impacts Endothelial Structure & Function. XOR is released by multiple sources, predominantly the liver. Upon release, XOR enters the vasculature and binds to the extracellular GAG-rich glycocalyx produced by endothelial cells. Inset: XOR can contribute to the breakdown of the glycocalyx initiate endothelial activation and promote intracellular ROS production. XOR can also be endocytosed following glycocalyx binding, which may impact endothelial cell function. Additionally, XOR functions to uncouple eNOS, inhibiting NO production, and instigating ONOO- formation. eNOS, endothelial nitric oxide synthase; GAG, glycosaminoglycan; ROS, reactive oxygen species; XOR, xanthine oxidoreductase; ONOO-, peroxynitrite.
Antioxidants 13 00712 g002
Figure 3. XOR’s Influence on Driving CKD. XOR directly contributes to CKD by increasing oxidative distress and the production of UA. Subsequently, XOR contributes to glomerular endothelial cell dysfunction, inflammation, and fibrosis. Genetic predisposition, environmental factors such as diet and heat, and aging can also contribute to total circulating XOR and renal pathogeneses. CVD, cardiovascular disease; GFR, glomerular filtration rate; MAFLD, metabolic-associated fatty liver disease; PKD, polycystic kidney disease; ROS, reactive oxygen species; UA, uric acid.
Figure 3. XOR’s Influence on Driving CKD. XOR directly contributes to CKD by increasing oxidative distress and the production of UA. Subsequently, XOR contributes to glomerular endothelial cell dysfunction, inflammation, and fibrosis. Genetic predisposition, environmental factors such as diet and heat, and aging can also contribute to total circulating XOR and renal pathogeneses. CVD, cardiovascular disease; GFR, glomerular filtration rate; MAFLD, metabolic-associated fatty liver disease; PKD, polycystic kidney disease; ROS, reactive oxygen species; UA, uric acid.
Antioxidants 13 00712 g003
Table 1. Selected preclinical and clinical studies of XOR inhibitors in CKD.
Table 1. Selected preclinical and clinical studies of XOR inhibitors in CKD.
Pre-Clinical Studies
DiseaseModel(s)Study DesignFindingsReference
DKDAkita mouse (genetic model of type 1 diabetes mellitus)Akita mice were treated with topiroxostat, a non-purine inhibitor of XOR for 4 weeks, and markers of XOR activity and diabetes-induced glomerular injury were measured.Compared to non-diabetic C57BL/6J controls, vehicle-treated Akita mice developed progressive diabetic glomerular injury, which was associated with increased XOR activity and oxidative stress. Pharmacological inhibition of XOR activity with topiroxostat markedly decreased renal ROS and progressive DKD in Akita mice.[7]
DKDdb/db mouse (model of obesity and type 2 diabetes mellitus)db/db mice were treated with various doses of XOR inhibitors (topiroxostat and febuxostat) for 4 weeks, and markers of systemic and renal XOR activity and kidney injury were assessed.Compared to control db/m littermates, control db/db mice exhibited increased systemic and intra-renal XOR activity, and this was associated with proteinuria. Treatment with topiroxostat significantly decreased XOR activity and oxidative stress in db/db mice. More importantly, topiroxostat rescued proteinuria in db/db mice in a dose-dependent fashion. While febuxostat significantly inhibited XOR activity in db/db mice, it did not significantly decrease proteinuria.[122]
DKDStreptozotocin-induced diabetic Sprague Dawley rat (chemically induced model of type 1 diabetes mellitus)Diabetic Sprague Dawley rats were treated with febuxostat for 7 weeks, and markers of XOR activity and renal oxidative stress, renal macrophage infiltration, and proteinuria were measured.In comparison to non-diabetic controls, diabetic Sprague Dawley rats exhibited increased XOR activity, renal inflammation, and renal injury, which were attenuated by treatment with febuxostat.[113]
DKDAkita, HFD-fed mice, STZ-induced diabetic B6-TG mice, and recombinant inbred BXD miceThis study employed multiomic and pharmacological approaches using the XOR inhibitors, allopurinol and febuxostat, to study the role of a XOR risk variant in the development of DKDThis study discovered that this identified variant is in the XOR promoter region and is a transcription factor binding site for C/EBPβ. Importantly, findings from this study demonstrated that the identified variants confer susceptibility to DKD.[5]
CKD
Non-DKD
5/6th nephrectomy Wistar rats with oxonic acid-induced hyperuricemia5/6th nephrectomized Wistar rats were made hyperuricemic with oxonic acid and treated with febuxostat for 4 weeks. Plasma uric acid, renal hemodynamics, proteinuria, and histopathologic evaluation were carried out in response to treatment with febuxostat.5/6th nephrectomized rats treated with oxonic acid displayed hyperuricemia, impaired renal hemodynamics and proteinuria versus vehicle-treated 5/6 nephrectomy rats. The administration of febuxostat decreased hyperuricemia, improved renal function, and ameliorated proteinuria in 5/6th nephrectomized rats that received oxonic acid.[123]
Obstructive nephropathyRat model of unilateral urethral obstructive (UUO) nephropathySprague Dawley rats were pretreated with febuxostat before UUO surgery and then daily after surgery for up to 14 days. Rats were sacrificed on days 1, 4, and 14 after surgery. Markers of intrarenal and systemic XOR activity, oxidative stress, renal inflammation, and tubulointerstitial fibrosis were assessed in response to treatment with febuxostat.Vehicle-treated UUO rats displayed increased XOR activity, renal inflammation, and tubulointerstitial fibrosis in comparison to sham rats. Pharmacological blockade of XOR with febuxostat rescued renal oxidative stress and inflammation and decreased tubulointerstitial fibrosis in UUO rats.[124]
Hypertension-induced CKDDahl salt-sensitive rat (genetic model of salt-sensitive hypertension and renal injury)Dahl salt-sensitive (SS) rats fed a high salt (8% NaCl) diet and treated with febuxostat daily for 4 or 8 weeks. XOR expression and XOR activity, oxidative stress, and progressive renal disease were measured in febuxostat-treated SS rats and their controls.Compared to normal salt (0.6% NaCl) diet-fed SS rats, high salt-diet-fed SS rats exhibited increased renal XOR expression and XOR activity, which was associated with oxidative stress and the progression of renal disease in SS rats. Treatment with febuxostat decreased XOR expression and activity and attenuated the progression of renal disease in high salt diet-fed SS rats.[125]
IgA NephropathygddY mouse (rodent model of IgA nephropathy)gddY mice either served as controls, or were treated with febuxostat for 9 weeks, during which XOR activity, inflammatory signaling, and the progression of renal disease were investigated.Control gddY mice demonstrated increased XOR activity, renal inflammation and fibrosis and renal disease versus BALB/c controls. XOR blockade with febuxostat was associated with decreased renal inflammation and renal disease in gddY mice.[126]
Hyperuricemic NephropathyAdenine + potassium oxonate-induced hyperuricemic Sprague Dawley rats.Control or febuxostat-treated hyperuricemic rats were treated for 5 weeks, and markers of XOR activity, endoplasmic reticulum stress, apoptosis, and renal disease were assessed.Treatment with febuxostat decreased XOR activity and renal urate deposition versus controls. Importantly, treatment with febuxostat ameliorated renal disease in hyperuricemic rats.[127]
Clinical Studies
DiseaseStudy PopulationStudy DesignOutcomesReference
DKDPatients with diabetes, concomitant with hyperuricemiaRandomized, double-blinded, placebo-controlled, and parallel study of patients with DKD and hyperuricemia. Recruited patients received either placebo or topiroxostat for 28 weeks. uACR was primary endpoint, while eGFR and serum uric acid levels were secondary endpoints.Topiroxostat decreased serum uric acid, while preventing renal function decline in the study population. However, topiroxostat did not have a significant effect on uACR in comparison to placebo.[119]
DKDDKD patients without goutCross sectional study of type 2 diabetes patients without a history of gout or allopurinol use. Serum creatinine and uric acid, HbA1C, and 24-hr urine protein excretion were measured.Serum uric acid showed a positive correlation with proteinuria in diabetic patients.[128]
DKDPatients with diabetes and asymptomatic hyperuricemiaRandomized, parallel-controlled trial that investigated the effects of allopurinol on renal function in diabetic patients and hyperuricemia.Effective control of serum uric acid with allopurinol was associated with decreased albumin excretion and the prevention on renal function decline in the study population.[129]
Hyperuricemic CKDHyperuricemic patients with stage 3 CKDRandomized, open label, parallel-group trial to investigate the renoprotective effects of febuxostat on hyperuricemic patients with stage 3 CKD.Treatment with febuxostat decreased serum uric acid and urinary levels of albumin and β2-macroglobulin in hyperuricemic CKD patients versus the control group. However, febuxostat did not influence markers of renal function (serum creatinine and eGFR).[130]
Hyperuricemic CKDHyperuricemic stage 3 CKD patients, with or without goutThis was a 22 week double-blind, multicentric, randomized trials that studied the safety and efficacy of topiroxostat in hyperuricemic patients with stage 3 CKD.Topiroxostat decreased serum urate and uACR in hyperuricemic stage 3 CKD patients versus placebo group. The observed effect of topiroxostat on renal function (i.e., eGFR) was not statistically significant. Adverse events were generally comparable between topiroxostat and placebo groups.[131]
Hyperuricemic CKDHyperuricemic patients with stage 3–4 CKDRandomized controlled trial that examined the efficacy of febuxostat on markers of endothelial dysfunction and renal function in CKD patients with asymptomatic hyperuricemia.Compared to the control group, febuxostat reduced serum uric acid and preserved renal function. However, febuxostat did not significantly decrease albuminuria or markers of endothelial dysfunction versus controls.[132]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Korsmo, H.W.; Ekperikpe, U.S.; Daehn, I.S. Emerging Roles of Xanthine Oxidoreductase in Chronic Kidney Disease. Antioxidants 2024, 13, 712. https://doi.org/10.3390/antiox13060712

AMA Style

Korsmo HW, Ekperikpe US, Daehn IS. Emerging Roles of Xanthine Oxidoreductase in Chronic Kidney Disease. Antioxidants. 2024; 13(6):712. https://doi.org/10.3390/antiox13060712

Chicago/Turabian Style

Korsmo, Hunter W., Ubong S. Ekperikpe, and Ilse S. Daehn. 2024. "Emerging Roles of Xanthine Oxidoreductase in Chronic Kidney Disease" Antioxidants 13, no. 6: 712. https://doi.org/10.3390/antiox13060712

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop