You are on page 1of 8

See discussions, stats, and author profiles for this publication at: https://www.researchgate.

net/publication/279730979

Uric acid as a modulator of glucose and lipid metabolism

Article  in  Biochimie · June 2015


DOI: 10.1016/j.biochi.2015.06.025 · Source: PubMed

CITATIONS READS

41 1,692

3 authors, including:

William Lima Valéria Ernestânia Chaves


Federal University of Minas Gerais Federal University of São João del-Rei
31 PUBLICATIONS   87 CITATIONS    34 PUBLICATIONS   389 CITATIONS   

SEE PROFILE SEE PROFILE

All content following this page was uploaded by William Lima on 23 November 2017.

The user has requested enhancement of the downloaded file.


Biochimie 116 (2015) 17e23

Contents lists available at ScienceDirect

Biochimie
journal homepage: www.elsevier.com/locate/biochi

Mini-review

Uric acid as a modulator of glucose and lipid metabolism


William Gustavo Lima a, Maria Emília Soares Martins-Santos b,
^nia Chaves a, *
ria Ernesta
Vale
a ~o Joa
Laboratory of Physiology, Federal University of Sa ~o del-Rei, Divino
polis, Minas Gerais, Brazil
b
Metabolic Biochemistry, Federal University of Sa~o Joa
~o del-Rei, Divinopolis, Minas Gerais, Brazil

a r t i c l e i n f o a b s t r a c t

Article history: In humans, uric acid is the final oxidation product of purine catabolism. The serum uric acid level is based
Received 28 January 2015 on the balance between the absorption, production and excretion of purine. Uric acid is similarly pro-
Accepted 26 June 2015 duced in the liver, adipose tissue and muscle and is primarily excreted through the urinary tract. Several
Available online 29 June 2015
factors, including a high-fructose diet and the use of xenobiotics and alcohol, contribute to hyper-
uricaemia. Hyperuricaemia belongs to a cluster of metabolic and haemodynamic abnormalities, called
Keywords:
metabolic syndrome, characterised by abdominal obesity, glucose intolerance, insulin resistance, dysli-
Uric acid
pidaemia and hypertension. Hyperuricaemia reduction in the Pound mouse or fructose-fed rats, as well
Metabolic syndrome
Non-alcoholic fatty liver
as hyperuricaemia induction by uricase inhibition in rodents and studies using cell culture have sug-
Adipose tissue gested that uric acid plays an important role in the development of metabolic syndrome. These studies
Skeletal muscle have shown that high uric acid levels regulate the oxidative stress, inflammation and enzymes associated
with glucose and lipid metabolism, suggesting a mechanism for the impairment of metabolic homeo-
stasis. Humans lacking uricase, the enzyme responsible for uric acid degradation, are susceptible to these
effects. In this review, we summarise the current knowledge of the effects of uric acid on the regulation
of metabolism, primarily focusing on liver, adipose tissue and skeletal muscle.
© 2015 Elsevier B.V. and Socie te
 Française de Biochimie et Biologie Mole
culaire (SFBBM). All rights
reserved.

1. Introduction

Uric acid is a metabolite derived from the oxidation of purine


Abbreviations: ABCG2, ATP-binding cassette transporter 2; ACC, acetyl-CoA compounds [1]. Since 1981, uric acid has been considered as a
carboxylase; ADH, alcohol dehydrogenase; Akt, protein kinase B; AMP, adenosine powerful chemical antioxidant, present in human plasma in con-
5'-monophosphate; AMPK, AMP activated kinase; ATP, adenosine triphosphate; centrations much higher than ascorbate [2]. It was hypothesized
ChoRE, carbohydrate response elements; ChREBP, carbohydrate responsive element
that uric acid can provide an antioxidant defense in humans against
binding protein; ERK, extracellular signal-regulated kinase; FAS, fatty acid synthase;
G6P, glucose-6-phosphate; G6Pc, glucose-6-phosphatase; GLUT, glucose trans- oxidant- and free radicals-caused ageing and cancer, because uric
porter; GMP, guanosine 5'-monophosphate; HDL, high density lipoprotein; HOMA- acid protects erythrocyte membrane against lipid peroxidation and
IR, homeostasis model assessment of insulin resistance; IMP, inosine 5'-mono- lysis induced by t-butylhydroperoxide in concentrations below
phosphate; IRS, insulin receptor substrate; LDH, lactate dehydrogenase; MAPK, those normally found in plasma [2]. Uric acid scavenges free radicals
mitogen-activated protein kinase; MCP-1, monocyte chemotactic protein-1; MRP4,
multi drug resistance protein4; NAD, nicotinamide adenine dinucleotide; NEAC,
under hydrophilic conditions, thereby inhibiting lipid peroxidation
nonenzymatic antioxidant capacity; NO, nitric oxide; NPT, Naþ-phosphate on the lipid-aqueous boundary, but this antioxidation is only slight
cotransporter; OAT, organic anion transporter; PEPCK, phosphoenolpyruvate car- under lipophilic conditions [3]. In the presence of pre-formed lipid
boxykinase; PI 3-kinase, phosphatidylinositol 3-kinase; PPAR, peroxisome hydro-peroxides, uric acid might accelerate the copper-induced
proliferator-activated receptor; ROS, reactive oxygen species; SREBP, sterol
peroxidation of human LDL, even in the presence of endogenous
responsive element binding protein; SMCT, sodium-dependent monocarboxylic
acid transporters; TAG, triacylglycerol; TORC2, transducer of regulated CREB activity antioxidants [4]. Thus, in hydrophobic environments, uric acid loses
2; URAT1, urate-anion exchanger. antioxidant ability and becomes a strong pro-oxidant.
* Corresponding author. Avenida Sebastia ~o Gonçalves Coelho, 400, Chanadour, More recently, uric acid effect on the development of insulin
35.501-296, Divino polis, Minas Gerais, Brazil.
resistance, dyslipidaemia and hypertension, important signals of
E-mail address: valeria.chaves@gmail.com (V.E. Chaves).

http://dx.doi.org/10.1016/j.biochi.2015.06.025
0300-9084/© 2015 Elsevier B.V. and Socie  te
 Française de Biochimie et Biologie Mole
culaire (SFBBM). All rights reserved.
18 W.G. Lima et al. / Biochimie 116 (2015) 17e23

Table 1
Biochemistry and haemodinamics changes induced by fructose or uric acid.

Fructose Model

Hyperuricaemia, hypertriglyceridaemia, hypertension Rats [20,26]


Steatosis Rats [21]
Increase of lipogenesisa Hep G2 [29]
Inhibition of AMPK phosphorylationa Hep G2 [40]

Uric acid Model

Hypertriglyceridaemia, hypertriglycaemia, hypertension Humans [6,7], Rats [28]


Increase of lipid contentb Hep G2 [29]
Inhibition of aconitase Hep G2 [29]
Inhibition of Akt and IRS-1 phosphorylation Liver, adipose tissue and muscle of mice, HepG2 [49]
Stimulation of gluconeogenesis Hep G2 [9]
Inhibition of AMPK phosphorylation Hep G2 [9]
Stimulation of NADPH oxidase Hep G2 [29], 3T3-L1 adipocytes [34]
Inhibition of nitric oxide production 3T3-L1 adipocytes [34]
Stimulation of renin-angiotensin system 3T3-L1 adipocytes [58]
Decrease of adiponectin RNAm 3T3-L1 adipocytes, human subcutaneous adipocytes [27]
a
Uric acid dependent.
b
Fructokinase independent. [Reference].

metabolic syndrome, has been suggested by clinical and epidemi- dynamic equilibrium between filtration, reabsorption and tubular
ological studies [5e8] (Table 1). secretion. Approximately 90% of the filtrated urate is reabsorbed
Purine compounds can be derived from exogenous sources, through a transport system mediated by urate-anion exchanger
such as high protein diets, or endogenous sources, such as the (URAT) 1. This transmembrane protein reabsorbs urate and secretes
catabolism of genetic material. The degradation of purine de- anionic organic compounds, such as lactate, ketone bodies and
rivatives, such as adenosine 5'-monophosphate (AMP) and gua- xenobiotics, through countertransport (Fig. 1). These anionic
nosine 5'-monophosphate (GMP), generates precursors for the organic compounds are released into renal tubular cells through
synthesis of endogenous uric acid [1]. Uric acid synthesis is initi- sodium-dependent monocarboxylic acid transporters (SMCT), such
ated with the cleavage of phosphate groups from AMP and GMP as SMCT1/2. During secretion, urate enters the cell at the baso-
through 5’-nucleotidases, releasing adenosine and guanosine lateral membrane via exchange with alpha-ketoglutarate, mediated
molecules, respectively. Adenosine deaminase converts adenosine by OAT1 and OAT3, or through exchange with unknown anions via
into inosine, while guanosine is converted into free guanine. OAT2. At the apical membrane, urate is secreted via multi-drug
Inosine is subsequently hydrolysed to hypoxanthine by purine resistance protein4 (MRP4), adenosine triphosphate (ATP)-bind-
nucleoside phosphorylase. Xanthine oxidase catalyses the last two ing cassette transporter 2 (ABCG2), Naþ-phosphate cotransporter
steps of uric acid synthesis: the conversion of hypoxanthine into (NPT) 1, and/or NPT4 [1,17].
xanthine and uric acid. Guanine is directly converted into Serum uric acid levels are controlled through the balance be-
xanthine, which is subsequently converted into uric acid by tween urate synthesis and excretion. Several exogenous and
xanthine oxidase [1]. AMP deaminase also converts AMP into endogenous factors can impair this balance, inducing hyper-
inosine 5'-monophosphate (IMP), which is then converted into uricaemia [18]. The excessive consumption of fructose, primarily
inosine by nucleotidases [9,10]. Xanthine oxidase is competitively used in sweetened beverages, has been considered an important
inhibited through drugs, such as allopurinol and febuxostat, which inducer of hyperuricaemia (Fig. 1) [19,20]. Excess fructose increases
are used in the treatment of pathologies characterised by hyper- fructokinase activity through a positive feedback mechanism via
uricaemia [11]. the activation of carbohydrate responsive element binding protein
In most mammals, uric acid is subsequently converted into (ChREBP) [21]. Fructokinase is responsible for the metabolism of
allantoin by uricase; thus, serum uric acid levels are typically low fructose into fructose-1-phosphate, resulting in adenosine
[12]. However, between 8 and 12 million years in the Miocene triphosphate (ATP) depletion in the liver, and this reaction occurs
period, as a result of different point mutations, human ancestors rapidly and without any negative feedback [22]. The decrease in
and other modern primates lost the ability to express the uricase, intracellular phosphate stimulates AMP deaminase, which cataly-
and consequently serum uric acid levels have increased [12,13]. It ses the degradation of AMP to inosine monophosphate, resulting in
has been suggested [14] that the loss of uricase in these evolu- a significant increase in serum uric acid levels (Fig. 1) [9,10]. Fruc-
tionary ancestors might have amplified the effects of fructose to tose and uric acid have been associated with cardiometabolic dis-
enhance fat stores. These increased lipid reserves could be mobi- eases [21,23], thus the growing consumption of sweetened
lised in times of energy demand, e.g., during the migration through beverages in developed and developing countries must be
large territorial extensions. However, the loss of uricase also in- controlled. Phytocompounds have also been associated with
creases blood pressure in response to salt [15]. Previous studies hyperuricaemia. Methylxanthines, derived from coffee, might be
have shown that humans have maintained the set of genes demethylated and subsequently converted into uric acid through
encoding this enzyme in the liver, however two premature stop xanthine oxidase (Fig. 1). Flavonoids stimulate DNA degradation
codons do not allow its expression [14,15]. and inhibit uric acid excretion, thereby elevating serum uric acid
In the absence of uricase, uric acid is the excretion product of levels (Fig. 1) [24]. In addition, some drugs also inhibit uric acid
purine metabolism [1]. Uric acid is a weak acid with a high disso- excretion. Pyrazinamide, used in the treatment of tuberculosis, and
ciation constant (pKa 5.8). Sodium urate is predominantly (98%) niacin, used in the treatment of dyslipidaemia, activate URAT1
deprotonated at physiological pH in the bloodstream. Renal (70%) transporter, thereby stimulating the reabsorption of filtrated urate
and biliary (30%) systems are responsible for the removal of urate (Fig. 1) [1,17]. Alcohol also stimulates this carrier by increasing the
from the bloodstream [1,16,17]. Renal excretion depends on the amount of lactate (Fig. 1). In this case, the hepatic metabolism of the
W.G. Lima et al. / Biochimie 116 (2015) 17e23 19

Fig. 1. Schematic representation of the regulatory factors of synthesis and excretion of uric acid. Fructose and phytocompounds, such as methylxanthines and flavonoids, increase
the rate of the endogenous synthesis of uric acid. Alcohol metabolism increases the levels of NADH, stimulates the lactate dehydrogenase (LDH) and increases the lactate synthesis.
As some xenobiotics, lactate activates the URAT-1 carrier, reducing the excretion of uric acid. An increase in the uric acid synthesis or a decrease in the uric acid excretion can
contribute to hyperuricaemia. ADH, alcohol dehydrogenase; LDH, lactate dehydrogenase; URAT1, urate-anion exchanger.

alcohol dehydrogenase system reduces the coenzyme NADþ to metabolic syndrome, such as hypertension and hyper-
NADH, which stimulates the conversion of pyruvate to lactate triglyceridaemia [20,26]. Fructose-fed rats treated with allopurinol,
through lactate dehydrogenase [25]. The lactate anion, generated at febuxostat or benzbromarone show normalised plasma uric acid
physiological pH, stimulates the activity of URAT1 [1,17]. and insulin levels and decreased TAG levels and systolic blood
Normal serum uric acid levels for men and women are 7 and pressure [20,26]. The continued administration of fructose induces
6 mg dL1, respectively [7,8]. Defects in the balance between uric a progressive increase in TAG and insulin levels [26]. Recent studies
acid excretion and synthesis, as described above, induce hyper- have demonstrated that lowering uric acid with allopurinol im-
uricaemia (Fig. 1). Epidemiological studies have associated hyper- proves the insulin sensitivity and hypertension in Pound mice, two
uricaemia with metabolic syndrome. The National Health and important features of metabolic syndrome [27]. Chronic hyper-
Nutrition Examination Survey (NHANES), analysing data from a uricaemia, induced through oxonic acid administration, causes
cross-sectional study involving 1370 children in the US between metabolic alterations, including postprandial hyper-
the ages of 12e17 years (1999e2002), has demonstrated a signifi- triglyceridaemia, systemic and glomerular hypertension, and he-
cant association between elevated serum uric acid levels and the patic TAG accumulation [28]. These findings suggest that uric acid
increased prevalence of abdominal obesity, hypertriglyceridaemia plays a causal role in the development of metabolic syndrome, and
and hyperglycaemia [7]. In 2008, a clinical study involving 3518 humans are susceptible to these effects, reflecting a lack of uricase
Caucasian patients between the ages of 21 and 85 years from Albert expression [14]. In this review, we summarise the current knowl-
Einstein Hospital in Sa~o Paulo showed that serum uric acid levels edge of the effects of uric acid in the regulation of metabolism,
were associated with increased triacylglycerol (TAG)/high density primarily focusing on liver, adipose tissue and muscle.
lipoprotein (HDL) and hepatic steatosis, independently of metabolic
syndrome and obesity [6]. In addition, a prospective analysis of the 2. Liver
Framingham heart study involving 4883 patients and a cohort
study involving 4292 patients demonstrated that individuals with The evaluation of TAG levels and Oil Red-O staining in the hu-
hyperuricaemia developed diabetes mellitus type 2 at significant man hepatocyte cell line HepG2 demonstrated that the lipid con-
higher rate than subjects with normal serum uric acid levels [8]. In tent is significantly increased through uric acid in a dose-
addition, a systemic review and meta-analysis of prospective dependent manner (from 0 to 750 mmol L1), while the presence
cohort studies provided strong evidence that high levels of serum of the xanthine oxidase inhibitor, allopurinol (100 mM), blocks uric
uric acid are observed independent of other established risk factors, acid production and TAG accumulation [29]. Previous studies have
particularly metabolic syndrome components, for the development shown that lowering uric acid through allopurinol treatment re-
of type 2 diabetes [5]. This clinical evidence indicates that uric acid duces hepatic steatosis in several models, such as, ethanol treated-
might increase the risk for metabolic syndrome, and basic studies rats [30], high fat diet-fed Mongolian gerbils [31], fructose-fed rats
have confirmed these findings (Table 1). Rats administered a 60% [21], and the Pound mouse [29]. The Pound mouse is a model of
fructose diet for 4 weeks show hyperuricaemia and symptoms of prediabetes/metabolic syndrome (Charles River, Wilmington, MA)
20 W.G. Lima et al. / Biochimie 116 (2015) 17e23

manifesting fatty liver, obesity, insulin resistance, and hypertension selective effect of NO and derived species [38]. The intraperitoneal
in response to a leptin receptor mutation. administration of uric acid to ob/ob mice normalises the activity of
In HepG2, uric acid up-regulates fructokinase expression (Fig. 1) complexes I and V in the mitochondrial respiratory chain in he-
through the activation of the transcription factor ChREBP, leading to patocytes and significantly improves the activity of complexes II
the amplification of the lipogenic effects of fructose [21]. ChREBP is and III [39]. Thus, the reduced nitric oxide levels contribute to the
activated by high glucose, independent of insulin, and this tran- effects of uric acid on the stimulation of mitochondrial oxidative
scription factor is responsible for the transcriptional activation of stress.
lipogenic genes, such as acetyl-CoA carboxylase (ACC) and fatty acid In addition, the generation of uric acid also inhibits AMP-
synthase (FAS), via direct binding to carbohydrate response ele- activated kinase (AMPK) phosphorylation in fructose-exposed
ments (ChoRE) in the promoter [32]. Mice lacking fructokinase are HepG2 cells [40]. AMPK stimulates fat oxidation through the inhi-
also protected from developing metabolic syndrome when exposed bition of ACC1 activity and the transport of malonyl-CoA to the
to a high-fructose diet [33]. However, uric acid also significantly mitochondria [41] and through the activation of peroxisome
increases TAG accumulation, even in cells lacking fructokinase and proliferator-activated receptor (PPAR)-a and its downstream target
aldolase B enzyme expression, suggesting that uric acid might genes [42]. AMPK also modulates hepatic lipogenesis through
function independently of direct fructose metabolism [29]. Uric multiple mechanisms, including the inactivation of transcription
acid increases lipogenesis in HepG2 cells through the inhibition of factors, such as sterol responsive element binding protein (SREBP)-
mitochondrial aconitase activity and the activation of ATP-citrate 1c and -2 [43,44] and ChREBP [45], resulting in the inhibition of the
lyase [29]. The inhibition of aconitase activity has been associated transcription of lipogenic target genes FAS, ACC1 and stearoyl-CoA
with the significant accumulation of its substrate, citrate, in the desaturase [43,44]. Thus, the inhibition of the energy sensor pro-
cytosol. Citrate is also a substrate for ATP-citrate lyase, which tein AMPK might represent an important step in the development
converts citrate into acetyl-CoA. The inhibition of ATP-citrate lyase of non-alcoholic fatty liver [46e48].
with radicicol (10 mmol L1) induces an increase in cytoplasmic Uric acids have also been suggested to play a role in hyper-
citrate levels in response to uric acid and blocks TAG accumulation, glycaemia. Rats with uricase inhibition through oxonic acid
indicating that citrate-stimulated ATP-citrate lyase is involved in administration show an increased metabolic response to fructose,
uric acid-mediated TAG accumulation [29]. The inhibition of FAS with higher glycaemia, blood pressure and renal damage [28].
activity through Cys75 (10 mmol L1), which ultimately converts Hyperuricaemia in mice treated with oxonic acid inhibits protein
acetyl-CoA and malonyl-CoA into long-chain saturated fatty acids kinase B (Akt) phosphorylation (Ser 473) in response to insulin in
(palmitate), also prevents TAG accumulation in response to fructose the liver, without changes in total Akt content [49]. In vitro hyper-
or uric acid [29]. Thus, uric acid increases fat in hepatocytes uricaemia induces oxidative stress in HepG2 cells and inhibits in-
through the stimulation of mitochondrial oxidative stress, which sulin signalling, as demonstrated through the phosphorylation Akt
blocks aconitase and increases citrate, thereby stimulating fat and insulin receptor substrate (IRS)-1. However, the addition of an
synthesis [29]. In addition, the silencing of the NADPH oxidase antioxidant blocks this hyperuricaemia-induced insulin signalling
subunit NOX4 partially prevents the increase in intracellular TAG impairment [49]. Thus, the hyperuricaemia effect on liver insulin
levels in response to fructose or uric acid [29]. Fructose also inhibits resistance is induced through increased oxidative stress. In the
mitochondrial aconitase activity and activates ATP-citrate lyase liver, Akt inhibits glycogen breakdown and de novo glucose pro-
[29]. Although antioxidant properties of allopurinol suggest that duction and stimulates the storage of glucose as glycogen and the
this drug might have off-target effects, the prevention of fructose- conversion of excess glucose into lipid [50]. Previous studies have
induced lipogenesis is dependent on the reduction of uric acid demonstrated that uric acid directly affects hepatic gluconeogen-
levels, as exogenous uric acid restored the metabolic effects of esis. De novo glucose production is significantly reduced in HepG2
fructose [29]. Similar findings were observed in vivo using the cells expressing uricases from ancestral hominids compared with
Pound mouse, a metabolic syndrome model that presents hyper- control cells [9]. However, the addition of uric acid activates
uricaemia and non-alcoholic fatty liver, leptin receptor defects, gluconeogenesis in a dose-dependent manner, suggesting that the
increased visceral fat and insulin resistance. The reduction of uric mechanism whereby uricase blocks glucose production is mediated
acid using allopurinol (30 mg kg1 in drinking water) effectively through uric acid degradation [9]. Uric acid addition reduces AMPK
reduces hepatic uric acid, TAG and cholesterol content in these activation, induced through the introduction of uricase in HepG2
mice. These findings are consistent with a significant decrease in cells [9]. AMPK activation blocks hepatic gluconeogenesis, in part,
the activity of the NADPH oxidase subunit NOX4, evaluated by through the inhibition of the transcription rate-limiting enzymes
hydrogen peroxide production, in the mitochondria of Pound mice phosphoenolpyruvate carboxykinase (PEPCK) and glucose-6-
treated with allopurinol and with concomitant higher mitochon- phosphatase (G6Pc) [51]. The effects of uric acid on gluconeogen-
drial aconitase activity and reduced cytoplasmic citrate release esis are mediated through AMPK via the phosphorylation of
[29]. The effects of allopurinol reducting the activity of NADPH transducer of regulated CREB activity 2 (TORC2), as the levels of
oxidase in liver mitochondria are consistent with previous data TORC2 phosphorylation are increased in uricase-expressing cells
showing that uric acid in adipocytes stimulates NADPH oxidase compared with control cells [9]. This phosphorylation inhibits the
[34]. transport of TORC2 into the nucleus and the expression of PEPCK
Studies have shown that uric acid decreases nitric oxide (NO) and G6Pc.
production in adipocytes and endothelial cells [34e36]. NO, syn- These basic biochemistry studies provide evidence that uric acid
thesised via different NO synthase isoforms, elicits changes in O2 might increase the risk for hepatic fat accumulation and hepatic
consumption, energy-conservation processes and free-radical glucose production, two important signals of metabolic syndrome
production to regulate mitochondrial respiratory functions [37]. [5e8,52].
The long-term incubation of liver mitochondrial membranes with
NO results in the persistent inhibition of NADH:cytochrome c 3. Adipose tissue
reductase activity, observed as inhibited NADH:ubiquinone reduc-
tase (Complex I) activity, whereas succinate:cytochrome c reduc- Although several studies have demonstrated an increase in uric
tase activity, including Complex II and Complex III electron transfer, acid levels in obese individuals [53,54], it has only recently been
remains unaffected [38]. However, uric acid partially inhibits the demonstrated that adipose tissue has abundant xanthine
W.G. Lima et al. / Biochimie 116 (2015) 17e23 21

oxidoreductase activity, similar to the liver, small intestine and control levels [27]. These experiments suggest that the uric acid-
other organs [55]. Furthermore, mature adipocytes and adipose induced inhibition of adiponectin production is not mediated
tissue produce and secrete uric acid [55]. Thus, it is likely that this through redox-dependent signalling, but rather through a mecha-
tissue contains all the enzymes necessary for purine catabolism. nism involving PPAR-g [27]. Similarly, the level of adiponectin
Interestingly, the visceral fat area is positively associated with the mRNA is decreased in the epididymal adipose tissue of xanthine
serum uric acid levels, indicative of the hyperuricaemia observed in oxidoreductase heterozygous knockout mice [62]. It has been pre-
subjects with type 2 diabetes mellitus [53]. It has previously been viously suggested that xanthine oxidoreductase is required for the
demonstrated that xanthine oxidoreductase is located upstream of activation of PPAR-g [Cheung 2007]. However, xanthine oxidase
PPARg and regulates the activity of this protein, which regulates inhibition with allopurinol in the Pound mice increases adiponectin
adipogenesis [56]. In addition, obesity increases the mRNA mRNA and serum levels and decreases MCP-1 mRNA expression
expression and activity of xanthine oxidoreductase and uric acid and serum levels [27]. Additional experiments are needed to clarify
secretion from adipose tissue [55,56]. However, more recently, it the effect of uric acid, oxidative stress and xanthine oxidoreductase
was demonstrated that rats fed a high-fat diet, a model of morbid on the synthesis of inflammatory cytokines in adipose tissue.
obesity, have an increase in xanthine oxidoreductase activity in However, uric acid might be directly involved in the development
subcutaneous, but not in visceral adipose tissue [57]. of insulin resistance in adipose tissue. Recently, it has been
In adipocytes, uric acid induces oxidative stress, stimulating demonstrated that hyperuricaemia decreases the phospho-Akt
NADPH oxidase [34] and the renin-angiotensin system [58]. Soluble content in the adipose tissue of mice treated with oxonic acid,
uric acid (15 mg dL1 during 30 min) stimulates an increase in without changes in total Akt [49], thereby inhibiting insulin sig-
reactive oxygen species (ROS) production and NADPH oxidase ac- nalling. The phosphatidylinositol 3-kinase (PI 3-kinase) and Akt
tivity in mature adipocytes, but not in preadipocytes [34]. It has signalling cascades are major regulators of glucose transporter
recently been demonstrated that renin-angiotensin system in- (GLUT) 4 exocytosis mediated through small GTPases in adipocytes
hibitors, such as losartan or captopril, prevent the increase in ROS [63]. Previous studies have demonstrated that lowering uric acid
induced through uric acid [58]. In 3T3-L1 adipocytes, uric acid also improves insulin sensitivity in Pound mice [27]. Future studies
(5 mg dL1 for 48 h) increases ROS production and up-regulates the should evaluate in vivo glucose uptake and lipogenesis in adipose
expression of the proteins involved in the renin-angiotensin system tissue from hyperuricaemic rodents.
(angiotensinogen, angiotensin-converting enzyme-1, renin, angio-
tensin type 1 receptor and angiotensin type 2 receptor) and 4. Muscle
angiotensin II protein secretion [58]. The stimulation of NADPH
oxidase-dependent ROS through uric acid activates the mitogen- Elevated serum uric acid is also one of the best independent
activated protein (MAP) kinases p38 and extracellular signal- predictors of diabetes and typically precedes the development of
regulated kinase (ERK)1/2, decreases nitric oxide bioavailability, both insulin resistance and diabetes [5]. Obese subjects with high
and increases protein nitrosylation and lipid oxidation [34]. levels of uric acid had lower insulin sensitivity (40%), evidenced by
Oxidative stress in adipose tissue has been recently recognised as a homeostasis model assessment of insulin resistance (HOMA-IR),
major causative factor for obesity-related inflammation and insulin-stimulated glucose disposal, and lower levels of oxidative
metabolic syndrome [59]. Thus, the oxidative modifications stress (30%) markers, compared with obese subjects with normal
induced through uric acid contribute to adipose tissue dysfunction. uric acid concentration [64]. Previous, it was demonstrated that
Indeed, it has been proposed that hyperuricaemia might be decreasing uric acid levels in Pound mice with allopurinol during 8
partially responsible for the proinflammatory endocrine imbalance weeks significantly improves insulin resistance, confirmed through
in the adipose tissue, representing an underlying mechanism of the insulin tolerance tests [27]. Considering that muscle exerts an
low-grade inflammation and insulin resistance observed in sub- important role in glucose homeostasis via insulin, it is possible that
jects with metabolic syndrome [27]. In 3T3-L1 adipocytes, the high levels of uric acid could exert some direct effect on this tissue.
presence of uric acid (5 or 15 mg dL1) in incubation medium for 3 Tsushima [55] provided the first evidence that muscle have higher
days increases the mRNA expression of monocyte chemotactic xanthine oxidoreductase activity in ob/ob than in C57 mice. High-
protein-1 (MCP-1) and decreases the mRNA expression of adipo- fat diet also induces an increase in xanthine oxidoreductase activ-
nectin [27]. These effects are similar in human primary subcu- ity in the skeletal muscle from rats [57]. More recently, Zhu [49]
taneous adipocytes [27]. Clinical evidence has revealed a negative showed that hyperuricaemia induces a decrease in the phospho-
correlation between the levels of uric acid and adiponectin in the Akt content in the muscle of mice treated with oxonic acid,
serum [60]. A strong positive association between serum leptin and without changes in total Akt, thereby inhibiting insulin signalling.
uric acid has been demonstrated in both diabetic and healthy Studies about the role of uric acid in the lipid and glucose meta-
subjects [61]; however, this biochemical mechanism still needs bolism in muscle cells are limited, making the role of this metab-
further clarification. olite poorly understood in this cell type. Thus, it would be
Superoxide scavengers or inhibitors of NADPH oxidase interesting and necessary to conduct experiments using muscle
completely prevent the increase in MCP-1 mRNA induced through cells or tissues in the presence of uric acid to elucidate the direct
uric acid in adipocytes, suggesting that the urate-induced activa- connection between high levels of uric acid and insulin resistance
tion of MCP-1 expression and secretion in adipocytes is mediated in skeletal muscle.
through superoxide-dependent ROS, likely generated through
NADPH oxidase [27]. Indeed, xanthine oxidoreductase heterozy- 5. Concluding Remarks
gous knockout mice at 18 months of age show an increase in
oxidative stress levels, characterised by the production of malon- Xanthine oxidoreductase in mammalian organs is predomi-
dialdehyde and H2O2, accompanied by the increased expression of nantly a nicotinamide adenine dinucleotide (NAD)þ-dependent
MCP-1 mRNA in epididymal adipose tissue [62]. However, the effect dehydrogenase that can be converted into an oxidase either irre-
of uric acid on adiponectin production is not inhibited in the versibly through limited proteolysis or reversibly through the
presence of superoxide scavengers or inhibitors of NADPH oxidase, chemical or enzymatic oxidation of thiol groups [65e68]. During
while the addition of rosiglitazone in the incubation medium oxygen-dependent reactions, this enzyme generates ROS, such as
restored adiponectin mRNA expression and adiponectin levels to superoxide anion and hydrogen peroxide [65,69]. Thus, the
22 W.G. Lima et al. / Biochimie 116 (2015) 17e23

reduction of uric acid using allopurinol or febuxostat also decreases [12] A.C. Keebaugh, J.W. Thomas, The evolutionary fate of the genes encoding the
purine catabolic enzymes in hominoids, birds, and reptiles, Mol. Biol. Evol. 27
ROS generation, making it difficult to interpret these findings.
(2010) 1359e1369.
However, the administration of the uricosuric agent benzbromar- [13] C. Zhang, K. Fan, W. Zhang, R. Zhu, L. Zhang, D. Wei, Structure-based char-
one reduces the hyperuricaemia, hypertriglyceridemia, and acterization of canine-human chimeric uricases and its evolutionary impli-
hyperinsulinaemia observed in fructose-fed rats [20,70], suggesting cations, Biochimie 94 (2012) 1412e1420.
[14] J.T. Kratzer, M.A. Lanaspa, M.N. Murphy, C. Cicerchi, C.L. Graves, P.A. Tipton,
that uric acid is a modulator of glucose and lipid metabolism. In E.A. Ortlund, R.J. Johnson, E.A. Gaucher, Evolutionary history and metabolic
addition, the hyperuricaemia induced through the administration insights of ancient mammalian uricases, Proc. Natl. Acad. Sci. U. S. A. 111
of oxonic acid causes metabolic alterations, including postprandial (2014) 3763e3768.
[15] S. Watanabe, D.H. Kang, L. Feng, T. Nakagawa, J. Kanellis, H. Lan, M. Mazzali,
hypertriglyceridaemia, hypertension, hepatic TAG accumulation R.J. Johnson, Uric acid, hominoid evolution, and the pathogenesis of salt
and impairment in response to insulin in liver, adipose tissue and sensitivity, Hypertension 40 (2002) 355e360.
muscle [28,49]. We believed that in vivo estimative of lipogenesis, [16] F. Roch-Ramel, B. Guisan, Renal transport of urate in humans, News Physiol.
Sci. 14 (1999) 80e84.
gluconeogenesis, glycolysis and glucose uptake in liver, adipose [17] M.S. Lipkowitz, Regulation of uric acid excretion by the kidney, Curr. Rheu-
tissue and/or muscle are needed to firmly establish the metabolic matol. Rep. 14 (2012) 179e188.
effects of uric acid. [18] E.P. Oliveira, R.C. Burini, High plasma uric acid concentration: causes and
Consequences, Diabetol. Metab. Synd. 4 (2012) 12.
[19] Z. Khitan, D.H. Kim, Fructose: a key factor in the development of metabolic
syndrome and hypertension, J. Nutr. Metab. 2013 (2013) 1e12.
Author contributions [20] T. Nakagawa, H. Hu, S. Zharikov, K.R. Tuttle, R.A. Short, O. Glushakova,
X. Ouyang, D.I. Feig, E.R. Block, J. Herrera-Acosta, J.M. Patel, R.J. Johnson,
All authors contributed to the development, analysis and A causal role for uric acid in fructose-induced metabolic syndrome, Am. J.
Physiol. Ren. Physiol. 290 (2006) F625eF631.
drafting of this review article.
[21] M.A. Lanaspa, L.G. Sanchez-Lozada, C. Cicerchi, N. Li, C.A. Roncal-Jimenez,
T. Ishimoto, M. Le, G.E. Garcia, J.B. Thomas, C.J. Rivard, A. Andres-Hernando,
B. Hunter, G. Schreiner, B. Rodriguez-Iturbe, Y.Y. Sautin, R.J. Johnson, Uric acid
Conflicts of interest stimulates fructokinase and accelerates fructose metabolism in the develop-
ment of fatty liver, PLoS One 7 (2012) e47948.
The authors have reported no conflicts of interest. [22] G. van den Berghe, M. Bronfman, R. Vanneste, H.G. Hers, The mechanism of
adenosine triphosphate depletion in the liver after a load of fructose. A kinetic
study of liver adenylate deaminase, Biochem. J. 162 (1977) 601e609.
[23] S.S. Essawy, K.A. Abdel-Sater, A.A. Elbaz, Comparing the effects of inorganic
Acknowledgements nitrate and allopurinol in renovascular complications of metabolic syn-
drome in rats: role of nitric oxide and uric acid, Arch. Med. Sci. 10 (2013)
The authors would like to thank Renato He lios Migliorini (in 537e545.
[24] S.B. Lotito, B. Frei, Consumption of flavonoid-rich foods and increased plasma
memoriam) for being an exemplary scientist and professor. This antioxidant capacity in humans: cause, consequence, or epiphenomenon?
work was supported through funding from the Federal University Free Radic. Biol. Med. 41 (2006) 1727e1746.
of S~ ~o del-Rei. W.G.L. received a fellowship from the Fundaça
ao Joa ~o [25] T. Neogi, C. Chen, J. Niu, C. Chaisson, D.J. Hunter, Y. Zhang, Alcohol quantity
and type on risk of recurrent gout attacks: an internet-based case-crossover
de Amparo a  Pesquisa do estado de Minas Gerais (FAPEMIG).
study, Am. J. Med. 127 (2014) 311e318.
[26] L.G. Sanchez-Lozada, E. Tapia, P. Bautista-García, V. Soto, C. Avila-Casado,
I.P. Vega-Campos, T. Nakagawa, L. Zhao, M. Franco, R.J. Johnson, Effects of
References febuxostat on metabolic and renal alterations in rats with fructose-
induced metabolic syndrome, Am. J. Physiol. Ren. Physiol. 294 (2008)
[1] A.K. Mandal, D.B. Mount, The molecular physiology of uric acid homeostasis, F710eF718.
Annu. Rev. Physiol 77 (2015) 323e345. [27] W. Baldwin, S. McRae, G. Marek, D. Wymer, V. Pannu, C. Baylis, R.J. Johnson,
[2] B.N. Ames, R. Cathcart, E. Schwiers, P. Hochstein, Uric acid provides an anti- Y.Y. Sautin, Hyperuricemia as a mediator of the proinflammatory endocrine
oxidant defense in humans against oxidant- and radical-caused aging and imbalance in the adipose tissue in a murine model of the metabolic syndrome,
cancer: a hypothesis, Proc. Natl. Acad. Sci. U. S. A. 78 (1981) 6858e6862. Diabetes 60 (2011) 1258e1269.
[3] S. Muraoka, T. Miura, Inhibition by uric acid of free radicals that damage [28] E. Tapia, M. Cristo bal, F.E. García-Arroyo, V. Soto, F. Monroy-Sa nchez,
biological molecules, Pharmacol. Toxicol. 93 (2003) 284e289. U. Pacheco, M.A. Lanaspa, C.A. Roncal-Jime nez, D. Cruz-Robles, T. Ishimoto,
[4] M. Bagnati, C. Perugini, C. Cau, R. Bordone, E. Albano, G. Bellomo, When and M. Madero, R.J. Johnson, L.G. Sa nchez-Lozada, Synergistic effect of uricase
why a water-soluble antioxidant becomes pro-oxidant during copper-induced blockade plus physiological amounts of fructose-glucose on glomerular hy-
low-density lipoprotein oxidation: a study using uric acid, Biochem. J. 340 pertension and oxidative stress in rats, Am. J. Physiol. Ren. Physiol. 304 (2013)
(1999) 143e152. F727eF736.
[5] Q. Lv, X.F. Meng, F.F. He, S. Chen, H. Su, J. Xiong, P. Gao, X.J. Tian, J.S. Liu, [29] M.A. Lanaspa, L.G. Sanchez-Lozada, Y.J. Choi, C. Cicerchi, M. Kanbay,
Z.H. Zhu, K. Huang, C. Zhang, High serum uric acid and increased risk of type 2 C.A. Roncal-Jimenez, T. Ishimoto, N. Li, G. Marek, M. Duranay, G. Schreiner,
diabetes: a systemic review and meta-analysis of prospective cohort studies, B. Rodriguez-Iturbe, T. Nakagawa, D.H. Kang, Y.Y. Sautin, R.J. Johnson, Uric acid
PLoS One 8 (2013) e56864. induces hepatic steatosis by generation of mitochondrial oxidative stress:
[6] T. Keenan, M.J. Blaha, K. Nasir, M.G. Silverman, R. Tota-Maharaj, potential role in fructose-dependent and -independent fatty liver, J. Biol.
J.A.M. Carvalho, R.D. Conceiça ~o, R.S. Blumenthal, R.D. Santos, Relation of uric Chem. 287 (2012) 40732e40744.
acid to serum levels of high-sensitivity C-reactive protein, triglycerides, and [30] H. Kono, I. Rusyn, B.U. Bradford, H.D. Connor, R.P. Mason, R.G. Thurman,
high-density lipoprotein cholesterol and to hepatic steatosis, Am. J. Cardiol. Allopurinol prevents early alcohol-induced liver injury in rats, J. Pharmacol.
110 (2012) 1787e1792. Exp. Ther. 293 (2000) 296e303.
[7] E.S. Ford, C. LI, S. Cook, H.K. Choi, Serum concentrations of uric acid and the [31] C.F. Xu, C.H. Yu, L. Xu, X.Y. Sa, Y.M. Li, Hypouricemic therapy: a novel potential
metabolic syndrome among US children and adolescents, Circulation 115 therapeutic option for nonalcoholic fatty liver disease, Hepatology 52 (2010)
(2007) 2526e2532. 1865e1866.
[8] W. Rathmann, E. Funkhouser, A.R. Dyer, J.M. Roseman, Relations of hyper- [32] X. Xu, J.S. So, J.G. Park, A.H. Lee, Transcriptional control of hepatic lipid
uricemia with the various components of the insulin resistance syndrome in metabolism by SREBP and ChREBP, Semin. Liver Dis. 33 (2013) 301e311.
young black and white adults: the CARDIA study. Coronary Artery Risk [33] T. Ishimoto, M.A. Lanaspa, M.T. Le, G.E. Garcia, C.P. Diggle, P.S. Maclean,
Development in Young Adults, Ann. Epidemiol. 8 (1998) 250e261. M.R. Jackman, A. Asipu, C.A. Roncal-Jimenez, T. Kosugi, C.J. Rivard,
[9] C. Cicerchi, N. Li, J. Kratzer, G. Garcia, C.A. Roncal-Jimenez, K. Tanabe, S. Maruyama, B. Rodriguez-Iturbe, L.G. Sanchez-Lozada, D.T. Bonthron,
B. Hunter, C.J. Rivard, Y.Y. Sautin, E.A. Gaucher, R.J. Johnson, M.A. Lanaspa, Uric Y.Y. Sautin, R.J. Johnson, Opposing effects of fructokinase C and A isoforms on
acid-dependent inhibition of AMP kinase induces hepatic glucose production fructose-induced metabolic syndrome in mice, Proc. Natl. Acad. Sci. U. S. A.
in diabetes and starvation: evolutionary implications of the uricase loss in 109 (2012) 4320e4325.
hominids, FASEB J. 28 (2014) 3339e3350. [34] Y.Y. Sautin, T. Nakagawa, S. Zharikov, R.J. Johnson, Adverse effects of the
[10] R.J. Johnson, T. Nakagawa, L.G. Sanchez-Lozada, M. Shafiu, S. Sundaram, M. Le, classic antioxidant uric acid in adipocytes. NADPH oxidase mediated oxida-
T. Ishimoto, Y.Y. Sautin, M.A. Lanaspa, Sugar, uric acid, and the etiology of tive/nitrosative stress, Am. J. Physiol. Cell Physiol. 293 (2007) C584eC596.
diabetes and obesity, Diabetes 62 (2013) 3307e3315. [35] Y.J. Choi, Y. Yoon, K.Y. Lee, T.T. Hien, K.W. Kang, K.C. Kim, J. Lee, M.Y. Lee,
[11] M.E. Ernst, M.A. Fravel, Febuxostat: a selective xanthine-oxidase/xanthine- S.M. Lee, D.H. Kang, B.H. Lee, Uric acid induces endothelial dysfunction by
dehydrogenase inhibitor for the management of hyperuricemia in adults vascular insulin resistance associated with the impairment of nitric oxide
with gout, Clin. Ther. 31 (2009) 2503e2518. synthesis, FASEB J. 28 (2014) 3197e3204.
W.G. Lima et al. / Biochimie 116 (2015) 17e23 23

[36] S. Zharikov, K. Krotova, H. Hu, C. Baylis, R.J. Johnson, E.R. Block, J. Patel, Uric syndrome, and subclinical coronary atherosclerosis, Am. J. Hypertens. 20
acid decreases NO production and increases arginase activity in cultured (2007) 83e89.
pulmonary artery endothelial cells, Am. J. Physiol. Cell Physiol. 295 (2008) [53] T.H. Kim, S.S. Lee, J.H. Yoo, S.R. Kim, S.J. Yoo, H.C. Song, Y.S. Kim, E.J. Choi,
C1183eC1190. Y.K. Kim, The relationship between the regional abdominal adipose tissue
[37] A. Boveris, L.E. Costa, E. Cadenas, J.J. Poderoso, Regulation of mitochondrial distribution and the serum uric acid levels in people with type 2 diabetes
respiration by adenosine diphosphate, oxygen and nitric oxide, Methods mellitus, Diabetol. Metab. Syndr. 4 (2012), 3.
Enzymol. 301 (1999) 188e198. [54] H.K. Choi, E.S. Ford, Prevalence of the metabolic syndrome in individuals with
[38] N.A. Riobo , E. Clementi, M. Melani, A. Boveris, E. Cadenas, S. Moncada, hyperuricaemia, Am. J. Med. 120 (2007) 442e447.
J.J. Poderoso, Nitric oxide inhibits mitochondrial NADH:ubiquinone reduc- [55] Y. Tsushima, H. Nishizawa, Y. Tochino, H. Nakatsuji, R. Sekimoto, H. Nagao,
tase activity through peroxynitrite formation, Biochem. J. 359 (2001) T. Shirakura, K. Kato, K. Imaizumi, H. Takahashi, M. Tamura, N. Maeda,
139e145. T. Funahashi, I. Shimomura, Uric acid secretion from adipose tissue and its
[39] I. García-Ruiz, C. Rodríguez-Juan, T. Díaz-Sanjuan, P. del Hoyo, F. Colina, increase in obesity, J. Biol. Chem. 288 (2013) 27138e27149.
T. Mun ~ oz-Yagüe, J.A. Solís-Herruzo, Uric acid and anti-TNF antibody improve [56] K.J. Cheung, I. Tzameli, P. Pissios, I. Rovira, O. Gavrilova, T. Ohtsubo, Z. Chen,
mitochondrial dysfunction in ob/ob mice, Hepatology 44 (2006) 581e591. T. Finkel, J.S. Flier, J.M. Friedman, Xanthine oxidoreductase is a regulator of
[40] M.A. Lanaspa, C. Cicerchi, G. Garcia, N. Li, C.A. Roncal-Jimenez, C.J. Rivard, adipogenesis and PPARgamma activity, Cell Metab. 5 (2007) 115e128.
B. Hunter, A. Andre s-Hernando, T. Ishimoto, L.G. S anchez-Lozada, J. Thomas, [57] A. Oberbach, J. Neuhaus, N. Schlichting, J. Kugler, S. Baumann, H. Till, Sleeve
R.S. Hodges, C.T. Mant, R.J. Johnson, Counteracting roles of AMP deaminase gastrectomy reduces xanthine oxidase and uric acid in a rat model of morbid
and AMP kinase in the development of fatty liver, PLoS One 7 (2012) obesity, Surg. Obes. Relat. Dis. 10 (2014) 684e690.
e48801. [58] J.X. Zhang, Y.P. Zhang, Q.N. Wu, B. Chen, Uric acid induces oxidative stress via
[41] D.G. Hardie, J. Corton, Y.P. Ching, S.P. Davies, S. Hawley, Regulation of lipid an activation of the renin-angiotensin system in 3T3-L1 adipocytes, Endocrine
metabolism by the AMP-activated protein kinase, Biochem. Soc. Trans. 25 48 (2015) 135e142, http://dx.doi.org/10.1007/s12020-014-0239-5 [Epub
(1997) 1229e1231. ahead of print].
[42] E. Barroso, R. Rodriguez-Calvo, L. Serrano-Marco, A.M. Astudillo, J. Balsinde, [59] S. Furukawa, T. Fujita, M. Shimabukuro, M. Iwaki, Y. Yamada, Y. Nakajima,
X. Palomer, M. V azquez-Carrera, The PPARbeta/delta activator GW501516 O. Nakayama, M. Makishima, M. Matsuda, I. Shimomura, Increased oxidative
prevents the downregulation of AMPK caused by a high-fat diet in liver and stress in obesity and its impact on metabolic syndrome, J. Clin. Invest 114
amplifies the PGC1alpha-Lipin 1-PPARalpha pathway leading to increased (2004) 1752e1761.
fatty acid oxidation, Endocrinology 152 (2011) 1848e1859. [60] S. Tamba, H. Nishizawa, T. Funahashi, Y. Okauchi, T. Ogawa, M. Noguchi,
[43] E.J. Jung, S.W. Kwon, B.H. Jung, S.H. Oh, B.H. Lee, Role of the AMPK/SREBP-1 K. Fujita, M. Ryo, S. Kihara, H. Iwahashi, K. Yamagata, T. Nakamura,
pathway in the development of orotic acid-induced fatty liver, J. Lipid. Res. I. Shimomura, Y. Matsuzawa, Relationship between the serum uric acid level,
52 (2011) 1617e1625. visceral fat accumulation and serum adiponectin concentration in Japanese
[44] Y. Li, S. Xu, M.M. Mihaylova, B. Zheng, X. Hou, B. Jiang, O. Park, Z. Luo, E. Lefai, men, Intern. Med. 47 (2008) 1175e1180.
J.Y. Shyy, B. Gao, M. Wierzbicki, T.J. Verbeuren, R.J. Shaw, R.A. Cohen, M. Zang, [61] B. Fruehwald-Schultes, A. Peters, W. Kern, J. Beyer, A. Pfützner, Serum leptin is
AMPK phosphorylates and inhibits SREBP activity to attenuate hepatic stea- associated with serum uric acid concentrations in humans, Metabolism 48
tosis and atherosclerosis in diet-induced insulin-resistant mice, Cell Metab. 13 (1999) 677e680.
(2011) 376e388. [62] N. Murakami, T. Ohtsubo, Y. Kansui, K. Goto, H. Noguchi, Y. Haga,
[45] T. Kawaguchi, K. Osatomi, H. Yamashita, T. Kabashima, K. Uyeda, Mechanism Y. Nakabeppu, K. Matsumura, T. Kitazono, Mice heterozygous for the xanthine
for fatty acid ‘‘sparing’’ effect on glucose-induced transcription: regulation of oxidoreductase gene facilitate lipid accumulation in adipocytes, Arterioscler.
carbohydrate-responsive element-binding protein by AMP activated protein Thromb. Vasc. Biol. 34 (2014) 44e51.
kinase, J. Biol. Chem. 277 (2002) 3829e3835. [63] M. Ishiki, A. Klip, Mini review: recent developments in the regulation of
[46] G. Musso, R. Gambino, M. Cassader, Emerging molecular targets for the glucose transporter-4 traffic: new signals, locations, and partners, Endocri-
treatment of nonalcoholic fatty liver disease, Annu. Rev. Med. 61 (2010) nology 146 (2005) 5071e5078.
375e392. [64] E. Fabbrini, M. Serafini, I. Colic Baric, S.L. Hazen, S. Klein, Effect of plasma uric
[47] J.H. Ix, K. Sharma, Mechanisms linking obesity, chronic kidney disease, and acid on antioxidant capacity, oxidative stress, and insulin sensitivity in obese
fatty liver disease: the roles of fetuin-A, adiponectin, and AMPK, J. Am. Soc. subjects, Diabetes 63 (2014) 976e981.
Nephrol. 21 (2010) 406e412. [65] F. Stirpe, M. Ravaioli, M.G. Battelli, S. Musiani, G.L. Grazi, Xanthine oxidore-
[48] B. Viollet, M. Foretz, B. Guigas, S. Horman, R. Dentin, L. Bertrand, L. Hue, ductase activity in human liver disease, Am. J. Gastroenterol. 97 (2002)
F. Andreelli, Activation of AMP-activated protein kinase in the liver: a new 2079e2085.
strategy for the management of metabolic hepatic disorders, J. Physiol. 574 [66] M.G. Battelli, Enzymic conversion of rat liver xanthine oxidase from dehy-
(2006) 41e53. drogenase (D form) to oxidase (O form), FEBS Lett. 113 (1980) 47e51.
[49] Y. Zhu, Y. Hu, T. Huang, Y. Zhang, Z. Li, C. Luo, Y. Luo, H. Yuan, I. Hisatome, [67] E. Della Corte, F. Stirpe, The regulation of rat liver xanthine oxidase.
T. Yamamoto, J. Cheng, High uric acid directly inhibits insulin signalling and Involvement of thiol groups in the conversion of the enzyme activity from
induces insulin resistance, Biochem. Biophys. Res. Commun. 447 (2014) dehydrogenase (type D) into oxidase (type O) and purification of the enzyme,,
707e714. Biochem. J. 126 (1972) 739e745.
[50] R. Hage Hassan, O. Bourron, E. Hajduch, Defect of insulin signal in peripheral [68] F. Stirpe, E. Della Corte, The regulation of rat liver xanthine oxidase. Conver-
tissues: Important role of ceramide, World J. Diabetes 5 (2014) 244e257. sion in vitro of the enzyme activity from dehydrogenase (type D) to oxidase
[51] P.A. Lochhead, I.P. Salt, K.S. Walker, D.G. Hardie, Sutherland C. 5- (type O), J. Biol. Chem. 244 (1969) 3855e3863.
aminoimidazole-4-carboxamide riboside mimics the effects of insulin on the [69] E.M. Link, P.A. Riley, Role of hydrogen peroxide in the cytotoxicity of the
expression of the 2 key gluconeogenic genes PEPCK and glucose-6- xanthine/xanthine oxidase system, Biochem. J. 249 (1988) 391e399.
phosphatase, Diabetes 49 (2000) 896e903. [70] U. Gresser, B.S. Gathof, M. Gross, Benzbromarone and fenofibrate are lipid
[52] T. de A. Coutinho, S.T. Turner, P.A. Peyser, L.F. Bielak, P.F. Sheedy 2nd, I.J. Kullo, lowering and uricosuric: a possible key to metabolic syndrome? Adv. Exp.
Associations of serum uric acid with markers of inflammation, metabolic Med. Biol. 370 (1994) 87e90.

View publication stats

You might also like