ArticlePDF Available

Isoorientin exerts a urate-lowering effect through inhibition of xanthine oxidase and regulation of the TLR4-NLRP3 inflammasome signaling pathway

Authors:

Abstract and Figures

Isoorientin (ISO), a natural flavonoid compound, has been identified in several plants and its biological activity is determined and the study on lowering uric acid has not been reported. In view of the current status of treatment of hyperuricemia, we evaluated the hypouricemic effects of ISO in vivo and in vitro, and explored the underlying mechanisms. Yeast extract-induced hyperuricemia animal model as well as hypoxanthine and xanthine oxidase (XOD) co-induced high uric acid L-O2 cell model and enzymatic experiments in vitro were selected. The XOD activity and uric acid (UA) level were inhibited after the treatment of ISO in vitro and in vivo. Furthermore, serum creatinine (CRE) and blood urea nitrogen (BUN) levels were also significantly reduced and liver damage was recovered in pathological histology after the ISO administration in hyperuricemia animal model. The results of mechanism illustrated that protein expressions such as XOD, toll-like receptor 4 (TLR4), cathepsin B (CTSB), NLRP3, and its downstream caspase-1 as well as interleukin-18 (IL-18) were markedly downregulated by ISO intervention in vitro and in vivo. Our results suggest that ISO exerts a urate-lowering effect through inhibiting XOD activity and regulating TLR4-NLRP3 inflammasome signal pathway, thus representing a promising candidate therapeutic agent for hyperuricemia. Both animal models and in vitro experiments suggested that ISO may effectively lower uric acid produce. The mechanism might be the inhibition of XOD activity and NLRP3 inflammasome of upregulation.
Content may be subject to copyright.
Vol.:(0123456789)
1 3
Journal of Natural Medicines
https://doi.org/10.1007/s11418-020-01464-z
ORIGINAL PAPER
Isoorientin exerts aurate‑lowering eect throughinhibition
ofxanthine oxidase andregulation oftheTLR4‑NLRP3 inammasome
signaling pathway
Meng‑FeiAn1,3· Ming‑YueWang1,3· ChangShen1,3· Ze‑RuiSun1,3· Yun‑LiZhao1,2,4· Xuan‑JunWang1,2,5·
JunSheng1,2,5
Received: 21 May 2020 / Accepted: 29 October 2020
© The Japanese Society of Pharmacognosy 2020
Abstract
Isoorientin (ISO), a natural flavonoid compound, has been identified in several plants and its biological activity is determined
and the study on lowering uric acid has not been reported. In view of the current status of treatment of hyperuricemia, we
evaluated the hypouricemic effects of ISO invivo and invitro, and explored the underlying mechanisms. Yeast extract-
induced hyperuricemia animal model as well as hypoxanthine and xanthine oxidase (XOD) co-induced high uric acid L-O2
cell model and enzymatic experiments invitro were selected. The XOD activity and uric acid (UA) level were inhibited
after the treatment of ISO invitro and invivo. Furthermore, serum creatinine (CRE) and blood urea nitrogen (BUN) levels
were also significantly reduced and liver damage was recovered in pathological histology after the ISO administration in
hyperuricemia animal model. The results of mechanism illustrated that protein expressions such as XOD, toll-like receptor
4 (TLR4), cathepsin B (CTSB), NLRP3, and its downstream caspase-1 as well as interleukin-18 (IL-18) were markedly
downregulated by ISO intervention invitro and invivo. Our results suggest that ISO exerts a urate-lowering effect through
inhibiting XOD activity and regulating TLR4-NLRP3 inflammasome signal pathway, thus representing a promising candidate
therapeutic agent for hyperuricemia.
Meng-Fei An and Ming-Yue Wang have contributed equally to this
work.
* Yun-Li Zhao
zhaoyunli123@163.com
* Xuan-Jun Wang
wangxuanjun@gmail.com
* Jun Sheng
shengj@ynau.edu.cn
1 Key Laboratory ofPu-erh Tea Science, Ministry
ofEducation, Yunnan Agricultural University,
Kunming650224, People’sRepublicofChina
2 College ofScience, Yunnan Agricultural University,
Kunming650224, People’sRepublicofChina
3 College ofFood Science andTechnology, Yunnan
Agricultural University, Kunming650224,
People’sRepublicofChina
4 Key Laboratory ofMedicinal Chemistry forNatural
Resource, Ministry ofEducation andYunnan Province,
School ofChemical Science andTechnology, Yunnan
University, Kunming650091, People’sRepublicofChina
5 State Key Laboratory forConservation andUtilization
ofBio-Resources inYunnan, Kunming650224,
People’sRepublicofChina
Journal of Natural Medicines
1 3
Graphic abstract
Both animal models and invitro experiments suggested that ISO may effectively lower uric acid produce. The mechanism
might be the inhibition of XOD activity and NLRP3 inflammasome of upregulation.
Keywords Isoorientin· Hyperuricemia· Xanthine oxidase· NLRP3 inflammasome· Uric acid
Abbreviations
ISO Isoorientin
XOD Xanthine oxidase
UA Uric acid
CRE Creatinine
BUN Blood urea nitrogen
TLR 4 Toll-like receptor 4
NF-κB Nuclear factor-κB
CTSB Cathepsin B
NLRP NOD-like receptor superfamily pyrin
PRRs Pattern recognition receptors
NLRC NOD-like receptor subfamily C
NAIP NLR family, apoptosis inhibitory protein
CARD Caspase activation and recruitment domain
PYHIN Pyrin and HIN domain-containing protein
URAT1 Urate anion transporter 1
GLUT9 Glucose transporter 9
OAT Organic cation transporter
ABCG2 ATP-binding cassette transporter ABCG2/
BCRP
ASC Apoptosis-associated speck-like protein
IL-1β Interleukin-1β
IL-18 Interleukin-18
IR Insulin resistance
LPS Lipopolysaccharide
K Potassium
Ca Calcium
FDA Food and Drug Administration
BCA Bicinchoninic acid
SPF Specific pathogen free
PBS Phosphate-buffered saline
PMSF Phenylmethylsulfonyl fluoride
DMSO Dimethyl sulfoxide
FBS Fetal bovine serum
MTT 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetra-
zolium bromide
DMEM Dulbecco’s modified Eagle’s medium
ROS Reactive oxygen species
H&E Hematoxylin and eosin
TGF-β Transforming growth factor beta
RIPA Radioimmunoprecipitation
PVDF Polyvinylidene fluoride
Introduction
Hyperuricemia is characterized by increased level of
serum uric acid exceeding 339μmol/L for women and
416μmol/L for men [1, 2]. The prevalence of hyperurice-
mia has increased annually worldwide over the past 2 dec-
ades [3], recorded as > 21% in USA [4], > 10% in Korea [5],
and > 13% in China [6].
Journal of Natural Medicines
1 3
Uric acid, the end product of purine metabolism in
humans, is generated by oxidation of xanthine oxidase.
About 90% urate is filtered by the kidney every day and
uric acid is reabsorbed through the sequential activities of
various transporters in the proximal tubules of the kidney
while the remainder is broken down by the small intestine
[7]. These transporters include the urate anion transporter 1
(URAT1) [8], glucose transporter 9 (GLUT9), organic cation
transporter (OAT) [9], and ATP-binding cassette transporter
ABCG2/BCRP (ABCG2) [10]. The majority of studies to
date have focused on URAT1, ABCG2, and GLUT9. In
rodents (rats and mice), uricase (uric acid oxidase) further
oxidizes uric acid to allantoin, which is more water soluble
with higher excretion efficiency from urine [11]. However,
humans and advanced primates lack a functional uricase
gene owing to gene mutations during the evolutionary pro-
cess [1]. Therefore, excessive production and limited excre-
tion of uric acid play an important role in the strong associa-
tions between hyperuricemia and disease.
Xanthine oxidase, mainly found in the liver, is the key
rate-limiting enzyme in the uric acid synthesis pathway,
which oxidizes hypoxanthine to xanthine and xanthine
further to uric acid. Accumulating evidence suggests that
high expression of XOD is correlated with blocking the
metabolism of thiopurine in Crohn’s disease [12], age in
vascular aging studies [13], pro-inflammatory responses in
heart failure [14], and insulin resistance in diabetes [15].
Elevated uric acid triggers the production and release of
various inflammatory mediators and cytokines in the body.
Moreover, increased xanthine oxidase activity and uric acid
promote activation of the NLRP3 inflammasome, in turn,
leading to stimulation of the downstream inflammatory fac-
tor, interleukin-1β (IL-1β), and release of interleukin-18
(IL-18) [16].
As part of the innate immune system, inflammasomes are
macromolecular polyprotein complexes recruited by pattern
recognition receptors to identify pathogenic microorganisms
and endogenous risk signals, i.e., pathogen-associated and
risk-related molecular patterns, which can activate cas-
pase-1, cleave IL-1β and IL-18 precursors to generate the
corresponding mature cytokines, and participate in clear-
ance of pathogens and adaptive immune responses of the
body [16]. In particular, the NLRP3 inflammasome, which
recognizes numerous pathogen types, has been extensively
characterized. The NLRP3 inflammasome corpuscle is
composed of NLRP3, apoptosis-associated speck-like
protein (ASC) containing a caspase activation and recruit-
ment domain (CARD), and pro-caspase-1 [17]. Three main
NLRP3 inflammasome activation pathways have been iden-
tified to date: (1) potassium ion (K+) efflux [18]; (2) lyso-
somal destruction [19, 20]; and (3) mitochondrial reactive
oxygen species (ROS) production [19]. Hyperuricemia is a
contributory factor for gout. Clinical studies have shown that
gout occurs due to deposition and stimulation of monoso-
dium urate (MSU) [21]. MSU is reported to bind the ubiq-
uitin domains of the three inflammasomes of the caspase-
1-activated NOD-like receptor superfamily, which triggers
the inflammatory response, producing active IL-1β and
IL-18 [22]. However, increasing evidence over recent years
suggests that elevated serum uric acid is accompanied by
inflammation, even in the absence of gout [23, 24]. Soluble
uric acid increases TLR-induced inflammatory factor acti-
vation in human primary cells [25] and amplifies human
intestinal cell inflammation through the activation of TLR4-
NLRP3 signaling pathway [26]. Furthermore, hyperuricemia
is accompanied by numerous health complications.
Hyperuricemia is regarded as a risk factor for several dis-
eases including gout, obesity, chronic kidney disease, and
diabetes [3, 27, 28]. At present, the treatment of hyperurice-
mia is mainly to inhibit uric acid synthesis drugs, promote
excretion of drugs, while inhibiting uric acid synthesis and
promoting excretion of drugs. Xanthine oxidase is the key
enzyme in uric acid synthesis. Inhibition of uric acid syn-
thesis has been successfully achieved through suppression
of xanthine oxidase activity. The xanthine oxidase inhibi-
tor, allopurinol, was approved by the US Food and Drug
Administration (FDA) in 1966 for treatment of recurrent
gouty arthritis, tophi, uric acid kidney stones, and other dis-
orders [1]. Importantly, however, allopurinol is commonly
associated with hypersensitivity reactions, including hepa-
totoxicity, Stevens–Johnson syndrome (SJS), toxic epider-
mal necrosis, eosinophilia, and systemic rash manifestations
[2931]. Febuxostat was subsequently approved by the Euro-
pean Medicines Agency and US FDA in 2008 for patients
intolerant to allopurinol [1]. Clinically, febuxostat has higher
urate-lowering activity and better safety than allopurinol
[32]. Initial studies have shown that the main adverse reac-
tions include joint pain, rash, diarrhea, and liver damage
[33, 34], most of which are mild to moderate and disappear
after termination of treatment [33, 35]. A series of complica-
tions caused by hyperuricemia, such as gout, the incidence is
increasing year by year; for the majority of patients, hyper-
uricemia is associated with extremely high costs of treat-
ment and poses a significant economic burden along with the
added burden of several side effects. Thus, identification of
active ingredients that lower uric acid level in natural plants
would not only prevent the occurrence of hyperuricemia
but also reduce the economic burden. Accordingly, natural
compounds that facilitate reduction of uric acid level have
become a hotspot of hyperuricemia research.
ISO, luteolin 6C-glucoside, is a flavonoid derived from
numerous plants, such as Commelina communis, Crataegus
pentagyna, Cymbopogon citratus, Cucumis satirus, Passi-
flora edulis, and Oxalis corniculata [36], with confirmed
biological activity [49]. The chemical structure of ISO was
shown in Fig.1a. Studies have shown that luteolin possess
Journal of Natural Medicines
1 3
significantly XOD inhibitory activity, antioxidant activity,
anti-inflammatory activity and so on [37]. And ISO is one
of the most common C-glycosides in luteolin [38]. Fur-
thermore, multiple biological properties of ISO have been
reported, including anti-cancer [3942], anti-oxidation [43,
44], anti-inflammatory [4547], anti-bacterial, anti-noci-
ceptive activity, and liver protection effects [45, 48]. For
instance, ISO has been shown to inhibit carbon tetrachloride
(CCl4)-induced hepatic fibrosis via free radical scavenging
and antioxidant activity, and regulation of nuclear factor-κB
(NF-κB) and transforming growth factor beta (TGF-β)/Smad
signaling pathways, and attenuate the activity of pro-inflam-
matory cytokines [49]. However, the study on lowering uric
acid has not been reported. Therefore, we hypothesized ISO
could regulate uric acid synthesis invivo and invitro, and
explored its mechanism primarily.
Materials andmethods
Reagents
Isoorientin (95–99%, BP0733/CAS No.: 4261-42-1) was
purchased from Chengdu Purifa Technology Development
Co., Ltd (Chengdu, China). In this study, xanthine oxidase
(CAS-9002-17-9), allopurinol (CAS-315-30-0), uric acid
(CAS-69-93-2), xanthine (CAS-69-89-6), and yeast extract
(2340951-02) were obtained from Sigma-Aldrich (St. Louis,
MO, USA), and hypoxanthine (H108384) was purchased
from Aladdin (Shanghai, China). The xanthine oxidase
(XOD) (A002) and uric acid (C012) test kits were purchased
from Nanjing Jiancheng Bioengineering Institute (Nanjing,
China), and 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetra-
zolium bromide (MTT) from Solarbio (Beijing, China).
Anti-NLRP3 antibody [EPR20425] (ab210491), anti-cas-
pase-1 antibody [EPR19672]-human, anti-IL-18 antibody
[EPR19954]-human, anti-IL-18 antibody [EPR19956]-
mouse, and anti-xanthine oxidase antibody [EPR4605]
(ab109235) were purchased from Abcam (Shanghai, China).
TLR4 (25): sc-293072-mouse was purchased from Santa
Cruz Biotechnology (Santa Cruz, CA, USA) and NF-κB
p65 (C22B4) rabbit mab was purchased from Cell Signaling
Technology (Beverly, MA, USA). Caspase-1 polyclonal anti-
body (Catalog No A0964) and cathepsin B polyclonal anti-
body (Catalog No A0967) were purchased from ABclonal
(Wuhan, China). In addition, anti-β-tubulin was purchased
from Proteintech Group, Inc. (Rosemont, IL, USA) while
horseradish peroxidase (HRP)-conjugated goat anti-mouse
IgG was purchased from R&D Systems (Minneapolis, MN,
USA).
In vitro assay ofxanthine oxidase activity
According to the related literatures [50], different con-
centrations of uric acid (i.e. 0mmol/L, 0.002 mmol/L,
0.004mmol/L, 0.006mmol/L, 0.01mmol/L, 0.02mmol/L,
0.04mmol/L, 0.08mmol/L, 0.10mmol/L) were diluted with
distilled water. The optical density (OD) values were meas-
ured at 295nm and adjusted to zero with distilled water to
obtain a standard curve of uric acid concentrations. Uric
acid (maximum absorption at the wavelength of 295nm)
is an indicator of xanthine oxidase activity. The uric acid
Fig. 1 Effect of ISO on XOD
activity in enzymatic analyses
invitro. a The chemical struc-
ture of ISO. b Establishment of
enzymatic experiments invitro.
c Effect of ISO on XOD activ-
ity. XOD, xanthine oxidase. The
applied concentration of allopu-
rinol was 80μmol/L invitro
enzymatic experiments. Data
were expressed as mean ± SEM
(n = 3) and representative of
three independent experiments.
Statistics: ###p < 0.001 versus
the control group; **p < 0.01
and ***p < 0.001 versus the
DMSO group
Journal of Natural Medicines
1 3
standard curve equation is y = Ax + B (y is the uric acid
content of the sample; x is the concentration of the sample
measured; A and B are coefficients of the equation). The
reaction system (300μL) was composed of 50μL test sam-
ple of various concentrations, 0.1mol/L Tris–HCL 170μL
(pH 7.6), 0.5mmol/L xanthine 30 μL, and 40 U/L xan-
thine oxidase (XOD) 50μL. All reaction components were
mixed in 96-well plate. At last, xanthine oxidase (XOD) was
added to initiate the reaction. The optical density at 295nm
(OD295) of the reaction system was monitored dynamically
for 20min at 25°C on a SpectraMax M3 microplate reader.
Enzyme activity (U) unit definition is the amount of
enzyme required to catalyze the conversion of 1μmol xan-
thine to uric acid in 1min at pH 7.6 and 25°C. XOD activity
and inhibition rate were calculated according to the follow-
ing formulae:
(A1 is the OD value measured before the reaction; A2 is the
OD value measured after the reaction; A and B are coeffi-
cients of the standard uric acid curve equation).
Urate‑lowering eect ofISO onhyperuricemic mice
induced byyeast extract
Specific pathogen-free (SPF) male ICR mice weighing
approximately 20–22g were purchased from Kunming
Medical University (license number SCXK 2015-0004). All
animals were housed at room temperature (20–25°C) and
constant humidity (40–70%) under a 12h light–dark cycle
in an SPF grade laboratory. Food and water were supplied
adlibitum. Animals were acclimated for 1week before the
experiment period. Animal studies were performed accord-
ing to the international guidelines of animal experiments
and internationally accepted ethical principles for labora-
tory animal use and care. The experiment was reviewed and
approved by the Institutional Animal Care and Use Commit-
tee of the Yunnan Agricultural University. (Animal ethics
approval code number is YUAN2019LLWYH003-2).
Mice were randomly divided into five groups: normal,
model, allopurinol (10mg/kg) treatment, ISO (10mg/kg)
treatment, and ISO (5mg/kg) treatment. The normal group
was intragastrically administered distilled water and the
other groups intragastrically administered yeast extract at a
dose of 15g/kg every day with reference to previous meth-
ods [51]. On the day of the experiment, the positive control
was administered intragastrically at 10mg/kg according to
the previous report [52], while treatments in the remaining
groups were administered via continuous intraperitoneal
Enzyme activity =(
A
2
A
1
B
)(
A
×min (20)) (UL),
Inhibition rate (%)=(standard tube enzyme activity test tube enzyme activity)standard tube enzyme activity ×100%,
injection. Treatments (allopurinol and ISO) were adminis-
tered after 6h. After 14days, mice were subjected to fast-
ing for 10h and blood collected via the orbital vein. Serum
samples were obtained by centrifuging blood at 4°C and
4000 × g for 10min, and serum UA, CRE, and BUN levels
determined using the Beckman AU680 automatic biochem-
istry analyzer (Kraemer Boulevard Brea, USA). Liver XOD
activity was measured using the xanthine oxidase (XOD)
test kit.
Histopathological examination
Tissues of the left upper lobe liver were embedded in paraf-
fin, cut into sections of 5μm thickness, and stained with
Hematoxylin and Eosin (H&E) for evaluation of general
morphology under a light microscope, as reported previ-
ously [53].
Determination ofuric acid level andXOD activity
incells
Normal liver cell line (L-O2 cells; MXC207) was purchased
from Shanghai Meixuan Biotechnology Co. Human normal
liver cell line (L-O2) was cultured in Dulbecco’s modified
Eagle’s medium (DMEM) supplemented with 10% fetal
bovine serum (FBS) and antibiotics (1% penicillin–strepto-
mycin) in 5% CO2 at 37°C.
To assess the inhibitory effects of ISO, L-O2 cells
were seeded in 96-well plates at a density of 5 × 104cells
per well and treated with different concentrations of ISO
(i.e. 25μmol/L, 50μmol/L, 100μmol/L, 200μmol/L, and
400μmol/L) for 24h, followed by addition of 20μL MTT.
After incubation for 4h in the dark, 200μL dimethyl sulfox-
ide (DMSO) was added to dissolve the remaining formazan
crystals of MTT. Following oscillation for 15min, cell via-
bility was measured at 492nm using a Flex Station 3 multi-
mode microplate reader (molecular devices).
L-O2 cells in the logarithmic growth phase were resus-
pended with DMEM and seeded in 6-well plates at a density
of 3 × 105cells per well. After a 24h inoculation period, the
supernatant in each well was discarded and gently washed
with phosphate-buffered saline (PBS) one time, followed by
the addition of 2mL ISO (i.e. 25μmol/L, 50μmol/L, and
100μmol/L) (ISO groups). The positive control allopurinol
concentration was set at 100μmol/L (prepared with complete
medium). After 24h of dosing, the supernatant in each well
was discarded. Cells were washed gently with PBS three
times and added 2mL hypoxanthine (800μmol/L). After
incubation for 24h at 37°C, xanthine oxidase was added at
Journal of Natural Medicines
1 3
a concentration of 4μL/well (final concentration is 0.3U/g),
incubation for 1h in dark, followed by extraction of superna-
tant or protein. Experiments were repeated at least three times.
The uric acid content of supernatant was measured using
a specific uric acid test kit (Nanjing, China). XOD activity
was measured using xanthine oxidase (XOD) test kit (Nanjing,
China).
Western blot analysis ofprotein contents incells
andliver tissues
After pre-treatment, cells were rinsed once with pre-cooled
PBS and treated with high-efficiency radioimmunoprecipita-
tion (RIPA) tissue/cell lysate (lysate: phenylmethylsulfonyl
fluoride (PMSF) = 100:1; 150μL/well). Cells were placed on
ice for 20min, transferred to 1.5mL centrifuge tubes, and
centrifuged at 4°C and 15,000 × g for 10min. Proteins from
liver tissues were extracted using RIPA tissue/cell lysates,
homogenized for 2min and centrifuged at 1036 × g for 10min
at 4°C. The supernatant fractions from cells and liver tissues
were transferred to fully labeled 1.5mL centrifuge tubes. The
bicinchoninic acid (BCA) assay kit was employed to determine
protein concentrations for preparing samples for western blot
analysis. Protein samples were separated using 8–12% poly-
acrylamide gels and transferred to a solid-phase carrier poly-
vinylidene fluoride (PVDF) membrane for 1h. Next, the mem-
brane was blocked in Tris-buffered saline with 0.1% Tween-20
(TBST) + 5% milk for 1h to block non-binding sites. Then, the
membrane were incubated overnight with specific antibod-
ies in PBS, including XOD (1:1000), NLRP3 (1:1000), IL-18
(1:200), CTSB (1: 1000), caspase-1 (1:1000), TLR4 (1:200),
NF-κB p65 (1:1000), and β-tubulin (1:1000), washed three
times with TBST for 5min each, and subsequently incubated
with horseradish peroxidase (HRP)-conjugated goat anti-rabbit
IgG (1:5000) diluted with TBST for 1h at room temperature.
After three 5min washes with TBST, images were acquired
using a Pro-light HRP Chemiluminescence Kit (Tiangen Bio-
tech, Beijing, China) on a FluorChem E System (Protein Sim-
ple, Santa Clara, CA, USA).
Statistical analysis
Analyses were performed with GraphPad Prism 5 (Graph-
Pad Software, Inc., La Jolla, CA, USA) and data presented as
mean ± standard error (SEM). p values < 0.05 were considered
statistically significant in all cases.
Results
Inhibition ofXOD byISO invitro
Xanthine oxidase is a key rate-limiting enzyme in uric acid
synthesis. As shown in Fig.1c, ISO inhibited XOD activ-
ity in a dose-dependent manner by 42μmol/L, 83μmol/L,
167μmol/L, 333μmol/L, and 667μmol/L ISO induced
12%, 25%, 51%, 78%, and 96% inhibition ratio, respec-
tively. The inhibitory effect of the highest concentrations
of ISO (667μmol/L) examined was similar to that of
allopurinol (80μmol/L), with a calculated inhibition ratio
of 97% (Fig.1b). These results indicated that ISO was a
potential XOD inhibitor.
Eects ofISO onbiochemical indicators
inhyperuricemia model induced byyeast extract
As shown in Fig.2, serum UA, CRE, and BUN levels
and liver XOD activity of the yeast extract-induced hyper-
uricemia model were significantly higher than the corre-
sponding measurements in the normal group (p < 0.01).
The positive control group (allopurinol) showed a marked
decrease in serum UA and CRE levels and liver XOD
activity, compared with the model group (p < 0.05/0.01).
As expected, serum UA, CRE, and BUN contents were
markedly reduced after ISO treatment at doses of 5mg/
kg and 10mg/kg (p < 0.05/0.01) while liver XOD activity
was inhibited to a limited extent (p > 0.05). Our findings
suggested that ISO not only reduced the uric acid level
and XOD activity but also protected kidneys from damage
caused by high-purine diets.
Liver histopathological changes
As shown in Fig.3, the normal group showed no obvious
degeneration or necrosis. The liver was rosy, shiny, and elas-
tic in appearance and tissue structure was normal. Hepato-
cytes in the model group displayed obvious pathological
changes. The structure of the hepatic lobule was unclear
and significant hepatocyte vacuolar degeneration, accompa-
nied by inflammatory cell infiltration, was observed. Upon
treatment with ISO, hepatocyte vacuolar degeneration and
inflammatory cell infiltration were significantly improved
in a dose-dependent manner. A similar improvement in the
inflammatory cell infiltration and hepatocyte vacuolation of
liver tissues was observed in the positive control—allopu-
rinol group although it can inhibit the activity of liver drug
enzyme. Our findings collectively demonstrated that ISO
exerted a protective effect on livers of hyperuricemic mice.
Journal of Natural Medicines
1 3
ISO suppressed XOD protein expression
anddownregulated activation oftheNLRP3
inammasome invivo
As shown in Fig.4, the expressions of XOD (Fig.4b) and
NLRP3 inflammatory signaling proteins such as TLR4
(Fig.4c), NF-κB (Fig.4d), CTSB (Fig.4e), NLRP3 (Fig.4f),
caspase-1 (Fig.4g) and IL-18 (Fig.4h) were significantly
increased in liver tissues of mice induced by yeast extract
compared with the normal group (p < 0.05). Notably, all of
them were downregulated after the treatment of ISO for suc-
cessive 14days compared to the model group (p < 0.05).
The similar results were observed in allopurinol group, of
them, the downregulation of NLRP3 inflammatory signaling
protein was inferior to that of ISO-10mg/kg group.
ISO induced signicant decrease onuric acid level
andXOD activity inhigh uric acid cell model
In this study, we established a liver cell model of high
uric acid co-induced by hypoxanthine and XOD. The uric
acid level was significantly increased by 30-fold in the
model, compared to control (Fig.5a). The inhibitory effect
of ISO on L-O2 cells was presented in Fig.5b. Interest-
ingly, ISO exerted no obvious inhibition at a concentra-
tion of 200μmol/L while the uric acid level was signifi-
cantly decreased at both 50μmol/L and 100μmol/L ISO
(p < 0.01) with inhibitory ratios 9% and 14%, respectively.
The inhibitory effect of 100μmol/L ISO was comparable
to that of allopurinol at the same concentration.
ISO inhibited XOD andNLRP3 inammasome
proteins expression inhigh uric acid cell model
The expressions of related proteins were examined to
explore the mechanism of lowering uric acid of ISO
invitro. Expectedly, the upregulated expression of these
proteins including XOD, NF-κB p65, CTSB, NLRP3, cas-
pase-1, IL-18 were observed in L-O2 high uric acid cell
model compared the control group (p < 0.05, Fig.6). How-
ever, the treatment with ISO could suppress the upregu-
lated expressions of them compared with those of model
group (p < 0.05/0.01), which is superior to that of the posi-
tive control (allopurinol) and consistent with the results
of invivo.
Fig. 2 Effects of ISO on serum
parameters and liver XOD activ-
ity in hyperuricemic mice. a
Uric acid (UA). b Serum blood
urea nitrogen (BUN). c Serum
creatinine (CRE). d Liver XOD
activity. The dose of allopurinol
was 10mg/kg invivo experi-
ments. Data are expressed as
mean ± SEM (n = 10). Statistics:
##p < 0.01 versus the normal
group; *p < 0.05 and **p < 0.01
versus the model group
Journal of Natural Medicines
1 3
Discussion
Currently available treatment agents for hyperuricemia, such
as allopurinol, febuxostat and other drugs, are associated
with a number of side effects. Accumulating studies have
highlighted the utility of natural components of plant materi-
als in effectively reducing uric acid [54], which may provide
further potential treatment options for hyperuricemia.
Hyperuricemia and its complications have considerable
detrimental effects on quality of life. Several factors, such
as high-purine foods and high-fructose corn syrup bever-
ages, trigger uric acid imbalance in the body. XOD, the key
to producing uric acid, is an indicator of hyperuricemia.
XOD oxidation of xanthine to uric acid is accompanied by
the production of superoxide anions [55]. In patients with
hyperuricemia, excessive ROS leads to oxidative stress,
further increasing the risk of disease secondary to hyper-
uricemia including cardiovascular events [56]. In addition,
the increase of uric acid is accompanied by inflammatory
disease such as gout and nephritis [16]. Therefore, ingredi-
ents with anti-inflammatory activity or antioxidant activity
may have therapeutic value in delaying these pathogenic
processes. Data from invitro enzymatic experiments in
the current study demonstrated that ISO inhibited XOD
activity in a dose-dependent manner (Fig.1c). Therefore,
the experimental models used in this paper were mainly
referred to the previous literatures. The main innovation of
this study was to explore the mechanisms of ISO on uric
acid synthesis.
Several types of hyperuricemic mouse models have been
developed to date. For instance, hyperuricemia could be co-
induced by hypoxanthine/xanthine and potassium oxonate
[57, 58], potassium oxonate [59] or yeast extract alone.
High protein and purine levels are responsible for purine
metabolism disorders. Yeasts rich in protein, nucleotides,
and B vitamins are fully decomposed in the body increase
nitrogen-containing organic bases and the content of uric
acid precursors such as phosphoric acid, promoting the pro-
duction of uric acid, is the best inducer for the study of uric
acid anabolism [60]. Moreover, hyperuricemia can cause
liver and kidney dysfunction [61], and liver disease or even
liver failure has been reported in patients treated with allopu-
rinol [62]. In uric acid-related excretory disorders that lead
to a decline in renal function, serum creatinine and blood
urea nitrogen are important indicators reflecting glomerular
filtration function and renal failure.
The hyperuricemia model induced by yeast extract and
high uric acid cell model co-induced by hypoxanthine and
XOD were selected to examine the effects of ISO on the uric
acid synthesis pathway. An animal model of hyperuricemia
Fig. 3 Hepatoprotective effects of ISO on liver morphology in hyper-
uricemic mice. Liver sections subjected to H&E staining (n = 10) are
presented at a magnification of × 200. Scale bar, 100μm. ISO (10mg/
kg and 5mg/kg) and allopurinol (10mg/kg) were respectively admin-
istered to mice in addition to yeast extract treatment
Journal of Natural Medicines
1 3
was successfully generated via intragastric administration of
yeast extract for 14days. Following intraperitoneal injection
of ISO for 14days, a significant reduction in serum UA,
BUN, and CRE levels were observed. ISO clearly exerted
a urate-lowering effect and improved glomerular filtration
function, reduced the possibility of kidney failure (Fig.2).
The protective effects of ISO were further examined via his-
topathological examination of the liver. ISO induced a sig-
nificant improvement in patchy necrosis and inflammatory
cell infiltration, compared with the model group (Fig.3).
Our data confirmed a hepatoprotective effect of ISO in the
hyperuricemia model, consistent with previous reports of
beneficial effects of it on liver fibrosis caused by alcohol
[48].
To date, studies on hyperuricemia mainly focused on
XOD activity [57] and XOD protein expression not deter-
mined [63]. In this study, both the activity and protein
expression of XOD were suppressed by ISO treatment
in animal and cells model confirming uric-acid-lowering
effect of ISO primarily by inhibiting XOD. Uric acid is
known to promote activation of the NLRP3 inflamma-
some [64]. Inflammation is an immune response in the
body that accompanied by inflammatory processes under
conditions of hyperuricemia. In our studies, ISO effec-
tively induced downregulation of NLRP3 inflammasome
as well as the downstream signal, caspase-1, and inflam-
matory factor, IL-18 invivo (Fig.4) and invitro (Fig.6).
A variety of endogenous and exogenous stimuli promote
assembly of NLRP3 inflammasome corpuscles, such as
uric acid, ROS, lysosomal leakage, lipopolysaccharide
(LPS), K+ efflux, and calcium ion (Ca2+) signals [65].
In most cases, the NLRP3 inflammasome is activated
by a dual signal mode. In resting cells, the components
of NLRP3 inflammasome corpuscles (NLRP3, ASC,
Fig. 4 Effects of ISO on protein levels in liver tissues. This figure
includes immunotbloting strips of protein expressions (a) levels of
XOD (b), TLR4 (c), NF-κB p65 (d), cathepsin B (e), NLRP3 (f),
caspase-1 (g), IL-18 (h) and β-tubulin were determined via Western
Blotting quantified using the software Image J Launcher. The dose of
allopurinol was 10mg/kg invivo experiments. These data were from
three separate experiments and expressed as the mean ± SEM. Statis-
tics: #p < 0.05 and ##p < 0.01 versus the normal group; *p < 0.05 and
**p < 0.01 versus the model group
Journal of Natural Medicines
1 3
and pro-caspase-1) and their substrates (pro-IL-1β and
pro-IL-18) are undetectable level [66]. Binding of pat-
tern recognition receptors, such as toll-like receptor, to
their corresponding ligands, act as a preliminary signal
that mediates nuclear translocation of NF-κB, trigger-
ing NLRP3, pro-IL-1β, and pro-IL-18 transcription and
thereby upregulating expression [66]. The second signal
mediates assembly of the NLRP3 inflammasome, activa-
tion of caspase-1, and processing of pro-IL-1β and pro-
IL-18, ultimately resulting in secretion of mature IL-1β
and IL-18 [66]. Notably, the expression levels of TLR4,
NF-κB p65, CTSB and the downstream regulation pro-
tein NLRP3 inflammasome were downregulated by ISO
invivo (Fig.4) and invitro (Fig.6), demonstrating that
UA production was inhibited by interference on NLRP3
inflammasome signal pathway. Based on the findings of
the study, we will explore the effect of ISO on uric acid
excretion and complications such as gout and nephritis
syndrome. Moreover, a new botanical drug rich in ISO
used in lowering uric acid is worth developing in our
future study.
Conclusions
In summary, ISO not only inhibited XOD activity and uric
acid level invitro and invivo, but also protected kidney
and liver from damage induced by high-purine diet invivo.
It was by this dual mechanism of action primarily by inhib-
iting XOD and regulating of the TLR4-NLRP3 inflamma-
some signaling pathway, which directly led to urate low-
ering. This study validates the therapeutic effects of ISO
against hyperuricemia and provides valuable insights into
the underlying biological mechanisms.
Fig. 5 Establishment of the
high uric acid cell model (a)
and effects of ISO on L-O2
cell toxicity (b), XOD activity
(c) and uric acid (UA) level
(d). L-O2 cells were seeded
in six-well plates overnight,
incubated with 2mL of dif-
ferent concentrations of ISO
(i.e. 25μmol/L, 50μmol/L,
and 100μmol/L) or allopu-
rinol (100μmol/L) for 24h,
followed by incubation with
hypoxanthine (800μmol/L) for
24h and addition of xanthine
oxidase (4μL/well). Data were
expressed as means ± SEM
(n = 3) and representative of
three independent experiments.
Statistics: ###p < 0.001 versus
the control group; *p < 0.05
versus the DMSO group in B,
***p < 0.001 versus the model
group in a, c and d
Journal of Natural Medicines
1 3
Funding This work was supported by the Yunnan provincial key
programs of Yunnan Eco-friendly Food International Cooperation
Research Center project under Grant (2019ZG00904, 2019ZG00909),
and the Science and Technology Plan Project of Yunnan Province
(2018IA060).
Compliance with ethical standards
Conflict of interest The authors declare that they have no conflict of
interest.
References
1. Chen C, Lu JM, Yao Q (2016) Hyperuricemia-related diseases
and xanthine oxidoreductase (XOR) inhibitors: an overview.
Med Sci Monit 22:2501–2512
2. Wang H, Zhang H, Sun L, Guo W (2018) Roles of hyperuricemia
in metabolic syndrome and cardiac-kidney-vascular system dis-
eases. Am J Transl Res 10(9):2749–2763
3. Ali N, Perveen R, Rahman S, Mahmood S, Rahman S, Islam S,
Haque T, Sumon AH, Kathak RR, Molla NH, Islam F, Mohanto
NC, Nurunnabi SM, Ahmed S, Rahman M (2018) Prevalence
of hyperuricemia and the relationship between serum uric
Fig. 6 Effects of ISO on protein levels invitro. This figure includes
immunbloting strips of protein expressions (a) levels of XOD (b),
TLR4 (c), NF-κB p65 (d), cathepsin B (e), NLRP3 (f), caspase-1 (g),
IL-18 (h) and β-tubulin were determined via western blotting quan-
tified using the software Image J Launcher. These data were from
three separate experiments and expressed as the mean ± SEM. Sta-
tistics: #p < 0.05, ##p < 0.01 and ###p < 0.001 versus the control group;
*p < 0.05 and **p < 0.01 versus the model group
Journal of Natural Medicines
1 3
acid and obesity: a study on Bangladeshi adults. PLoS ONE
13(11):e0206850
4. Zhu Y, Pandya BJ, Choi HK (2011) Prevalence of gout and hyper-
uricemia in the US general population: the National Health and
Nutrition Examination Survey 2007–2008. Arthritis Rheumatol
63(10):3136–3141
5. Kim Y, Kang J, Kim GT (2018) Prevalence of hyperuricemia and
its associated factors in the general Korean population: an analy-
sis of a population-based nationally representative sample. Clin
Rheumatol 37(9):2529–2538
6. Liu R, Han C, Wu D, Xia X, Gu J, Guan H, Shan Z, Teng W
(2015) Prevalence of hyperuricemia and gout in Mainland China
from 2000 to 2014: a systematic review and meta-analysis.
Biomed Res Int 2015:762820
7. Tan PK, Farrar JE, Gaucher EA, Miner JN (2016) Coevolu-
tion of URAT1 and uricase during primate evolution: impli-
cations for serum urate homeostasis and gout. Mol Biol Evol
33(9):2193–2200
8. Tan PK, Ostertag TM, Miner JN (2016) Mechanism of high affin-
ity inhibition of the human urate transporter URAT1. Sci Rep
6:34995
9. Wang Z, Cui T, Ci X, Zhao F, Sun Y, Li Y, Liu R, Wu W, Yi X,
Liu C (2019) The effect of polymorphism of uric acid transporters
on uric acid transport. J Nephrol 32(2):177–187
10. Fujita K, Ichida K (2018) ABCG2 as a therapeutic target candidate
for gout. Expert Opin Ther Targets 22(2):123–129
11. Desideri G, Castaldo G, Lombardi A, Mussap M, Testa A, Pon-
tremoli R, Punzi L, Borghi C (2014) Is it time to revise the normal
range of serum uric acid levels? Eur Rev Med Pharmacol Sci
18(9):1295–1306
12. Ansari A, Aslam Z, De Sica A, Smith M, Gilshenan K, Fairbanks
L, Marinaki A, Sanderson J, Duley J (2008) Influence of xanthine
oxidase on thiopurine metabolism in Crohn’s disease. Aliment
Pharmacol Ther 28(6):749–757
13. Aranda R, Domenech E, Rus AD, Real JT, Sastre J, Vina J,
Pallardo FV (2007) Age-related increase in xanthine oxi-
dase activity in human plasma and rat tissues. Free Radic Res
41(11):1195–1200
14. Alem MM, Alshehri AM, Cahusac PM, Walters MR (2018)
Effect of xanthine oxidase inhibition on arterial stiffness in
patients with chronic heart failure. Clin Med Insights Cardiol
12:1179546818779584
15. El-Bassossy HM, Watson ML (2015) Xanthine oxidase inhibition
alleviates the cardiac complications of insulin resistance: effect
on low grade inflammation and the angiotensin system. J Transl
Med 13:82
16. Ives A, Nomura J, Martinon F, Roger T, LeRoy D, Miner JN,
Simon G, Busso N, So A (2015) Xanthine oxidoreductase regu-
lates macrophage IL1beta secretion upon NLRP3 inflammasome
activation. Nat Commun 6:6555
17. Bauernfeind F, Hornung V (2013) Of inflammasomes and patho-
gens–sensing of microbes by the inflammasome. EMBO Mol Med
5(6):814–826
18. Amores-Iniesta J, Barbera-Cremades M, Martinez CM, Pons JA,
Revilla-Nuin B, Martinez-Alarcon L, Di Virgilio F, Parrilla P,
Baroja-Mazo A, Pelegrin P (2017) Extracellular ATP activates
the NLRP3 inflammasome and is an early danger signal of skin
allograft rejection. Cell Rep 21(12):3414–3426
19. Bai H, Yang B, Yu W, Xiao Y, Yu D, Zhang Q (2018) Cathepsin B
links oxidative stress to the activation of NLRP3 inflammasome.
Exp Cell Res 362(1):180–187
20. Amaral EP, Riteau N, Moayeri M, Maier N, Mayer-Barber KD,
Pereira RM, Lage SL, Kubler A, Bishai WR, D’Imperio-Lima
MR, Sher A, Andrade BB (2018) Lysosomal cathepsin release is
required for NLRP3-inflammasome activation by Mycobacteri-
umtuberculosis in infected macrophages. Front Immunol 9:1427
21. Dalbeth N, Merriman TR, Stamp LK (2016) Gout. Lancet
388(10055):2039–2052
22. Liu-Bryan R, Pritzker K, Firestein GS, Terkeltaub R (2005) TLR2
signaling in chondrocytes drives calcium pyrophosphate dihydrate
and monosodium urate crystal-induced nitric oxide generation. J
Immunol 174(8):5016–5023
23. Ghaemi-Oskouie F, Shi Y (2011) The role of uric acid as an
endogenous danger signal in immunity and inflammation. Curr
Rheumatol Rep 13(2):160–166
24. Grainger R, McLaughlin RJ, Harrison AA, Harper JL (2013)
Hyperuricaemia elevates circulating CCL2 levels and primes
monocyte trafficking in subjects with inter-critical gout. Rheu-
matology 52(6):1018–1021
25. Crisan TO, Cleophas MC, Oosting M, Lemmers H, Toenhake-
Dijkstra H, Netea MG, Jansen TL, Joosten LA (2016) Soluble uric
acid primes TLR-induced proinflammatory cytokine production
by human primary cells via inhibition of IL-1Ra. Ann Rheum Dis
75(4):755–762
26. Chen M, Lu X, Lu C, Shen N, Jiang Y, Chen M, Wu H (2018)
Soluble uric acid increases PDZK1 and ABCG2 expression in
human intestinal cell lines via the TLR4-NLRP3 inflammasome
and PI3K/Akt signaling pathway. Arthritis Res Ther 20(1):20
27. Bakan A, Oral A, Elcioglu OC, Takir M, Kostek O, Ozkok A,
Basci S, Sumnu A, Ozturk S, Sipahioglu M, Turkmen A, Voro-
neanu L, Covic A, Kanbay M (2015) Hyperuricemia is associ-
ated with progression of IgA nephropathy. Int Urol Nephrol
47(4):673–678
28. Gromadzinski L, Januszko-Giergielewicz B, Pruszczyk P (2015)
Hyperuricemia is an independent predictive factor for left ventric-
ular diastolic dysfunction in patients with chronic kidney disease.
Adv Clin Exp Med 24(1):47–54
29. Lin MS, Dai YS, Pwu RF, Chen YH, Chang NC (2005) Risk
estimates for drugs suspected of being associated with Stevens-
Johnson syndrome and toxic epidermal necrolysis: a case-control
study. Intern Med J 35(3):188–190
30. Zemmez Y, Hjira N (2018) Allopurinol-induced DRESS syn-
drome: drug reaction with eosynophilia and systemic symptoms
(DRESS). Pan Afr Med J 30:120
31. Mockenhaupt M, Viboud C, Dunant A, Naldi L, Halevy S, Bou-
wes Bavinck JN, Sidoroff A, Schneck J, Roujeau JC, Flahault A
(2008) Stevens-Johnson syndrome and toxic epidermal necroly-
sis: assessment of medication risks with emphasis on recently
marketed drugs. The EuroSCAR-study. J Investig Dermatol
128(1):35–44
32. Chohan S (2011) Safety and efficacy of febuxostat treatment in
subjects with gout and severe allopurinol adverse reactions. J
Rheumatol 38(9):1957–1959
33. Yu KH (2007) Febuxostat: a novel non-purine selective inhibitor
of xanthine oxidase for the treatment of hyperuricemia in gout.
Recent Pat Inflamm Allergy Drug Discov 1(1):69–75
34. Bohm M, Vuppalanchi R, Chalasani N, Drug-Induced Liver
Injury N (2016) Febuxostat-induced acute liver injury. Hepatol-
ogy 63(3):1047–1049
35. Bruce SP (2006) Febuxostat: a selective xanthine oxidase inhibitor
for the treatment of hyperuricemia and gout. Ann Pharmacother
40(12):2187–2194
36. Lopez-Lazaro M (2009) Distribution and biological activities of
the flavonoid luteolin. Mini Rev Med Chem 9(1):31–59
37. Yan J, Zhang G, Hu Y, Ma Y (2013) Effect of luteolin on xanthine
oxidase: inhibition kinetics and interaction mechanism merging
with docking simulation. Food Chem 141(4):3766–3773
38. Pauff JM, Hille R (2009) Inhibition studies of bovine xanthine
oxidase by luteolin, silibinin, quercetin, and curcumin. J Nat Prod
72(4):725–731
39. Lin X, Wei J, Chen Y, He P, Lin J, Tan S, Nie J, Lu S, He M, Lu
Z, Huang Q (2016) Isoorientin from Gypsophila elegans induces
Journal of Natural Medicines
1 3
apoptosis in liver cancer cells via mitochondrial-mediated path-
way. J Ethnopharmacol 187:187–194
40. Yuan L, Wang J, Xiao H, Wu W, Wang Y, Liu X (2013) MAPK
signaling pathways regulate mitochondrial-mediated apoptosis
induced by isoorientin in human hepatoblastoma cancer cells.
Food Chem Toxicol 53:62–68
41. Yuan L, Wei S, Wang J, Liu X (2014) Isoorientin induces apop-
tosis and autophagy simultaneously by reactive oxygen species
(ROS)-related p53, PI3K/Akt, JNK, and p38 signaling pathways
in HepG2 cancer cells. J Agric Food Chem 62(23):5390–5400
42. Yuan L, Wang Y, Wang J, Xiao H, Liu X (2014) Additive effect
of zinc oxide nanoparticles and isoorientin on apoptosis in human
hepatoma cell line. Toxicol Lett 225(2):294–304
43. Da Silva MM, De Assis AM, Da Rocha RF, Gasparotto J, Gazola
AC, Costa GM, Zucolotto SM, Castellanos LH, Ramos FA,
Schenkel EP, Reginatto FH, Gelain DP, Moreira JC (2013) Pas-
siflora manicata (Juss.) aqueous leaf extract protects against reac-
tive oxygen species and protein glycation invitro and exvivo
models. Food Chem Toxicol 60:45–51
44. Yuan L, Wang J, Wu W, Liu Q, Liu X (2016) Effect of isoorientin
on intracellular antioxidant defence mechanisms in hepatoma and
liver cell lines. Biomed Pharmacother 81:356–362
45. Yuan L, Han X, Li W, Ren D, Yang X (2016) Isoorientin prevents
hyperlipidemia and liver injury by regulating lipid metabolism,
antioxidant capability, and inflammatory cytokine release in high-
fructose-fed mice. J Agric Food Chem 64(13):2682–2689
46. Yuan L, Wu Y, Ren X, Liu Q, Wang J, Liu X (2014) Isoorien-
tin attenuates lipopolysaccharide-induced pro-inflammatory
responses through down-regulation of ROS-related MAPK/NF-
kappaB signaling pathway in BV-2 microglia. Mol Cell Biochem
386(1–2):153–165
47. Lee W, Ku SK, Bae JS (2014) Vascular barrier protective effects
of orientin and isoorientin in LPS-induced inflammation invitro
and invivo. Vasc Pharmacol 62(1):13–14
48. Chen YX, Huang QF, Lin X, Wei JB (2013) Protective effect of
isoorientin on alcohol-induced hepatic fibrosis in rats. Zhongguo
Zhong Yao Za Zhi 38(21):3726–3730
49. Lin X, Chen Y, Lv S, Tan S, Zhang S, Huang R, Zhuo L, Liang
S, Lu Z, Huang Q (2015) Gypsophila elegans isoorientin attenu-
ates CCl(4)-induced hepatic fibrosis in rats via modulation of
NF-kappaB and TGF-beta1/Smad signaling pathways. Int Immu-
nopharmacol 28(1):305–312
50. Zhu SY, Zhou YD, Liu QS, Du GH (2007) Establishment and
application of a high-throughput screening assay for xanthine
oxidase inhibitor invitro. Chin Pharm J 42(3):187–190
51. Chen GL, Sun XX, Wang QM, Zhang XM, Liu PH (2001) Study
on hyperuricemia model in mice. Chin Pharm Bull 17(3):350–352
52. Niu YF, Liu K, Gao LH, Liu X, Li L (2015) Effects of 3,5,2’,4’-tet-
rahydroxychalcone on serum uric acid levels and the content of
hepatic XOD/XDH in mice. Chin Pharm J 50(1):34–38
53. Xiong X, Ren Y, Cui Y, Li R, Wang C, Zhang Y (2017) Obet-
icholic acid protects mice against lipopolysaccharide-induced liver
injury and inflammation. Biomed Pharmacother 96:1292–1298
54. Chen L, Li M, Wu JL, Li JX, Ma ZC (2019) Effect of lemon water
soluble extract on hyperuricemia in a mouse model. Food Funct
10(9):6000–6008
55. Hayyan M, Hashim MA, AlNashef IM (2016) Superox-
ide ion: generation and chemical implications. Chem Rev
116(5):3029–3085
56. Enroth C, Eger BT, Okamoto K, Nishino T, Nishino T, Pai EF
(2000) Crystal structures of bovine milk xanthine dehydrogenase
and xanthine oxidase: structure-based mechanism of conversion.
Proc Natl Acad Sci USA 97(20):10723–10728
57. Hou CW, Lee YC, Hung HF, Fu HW, Jeng KC (2012) Longan
seed extract reduces hyperuricemia via modulating urate trans-
porters and suppressing xanthine oxidase activity. Am J Chin Med
40(5):979–991
58. Qin Z, Wang S, Lin Y, Zhao Y, Yang S, Song J, Xie T, Tian J, Wu
S, Du G (2018) Antihyperuricemic effect of mangiferin aglycon
derivative J99745 by inhibiting xanthine oxidase activity and urate
transporter 1 expression in mice. Acta Pharm Sin B 8(2):306–315
59. Hongyan L, Suling W, Weina Z, Yajie Z, Jie R (2016) Antihy-
peruricemic effect of liquiritigenin in potassium oxonate-induced
hyperuricemic rats. Biomed Pharmacother 84:1930–1936
60. Shuyan Y, Mingsan M (2014) Interventional study of total flavone
extract of lysimashia on hyperuricemia animal models. Henan
University of Chinese Medicine, Zheng Zhou
61. Minami M, Ishiyama A, Takagi M, Omata M, Atarashi K (2005)
Effects of allopurinol, a xanthine oxidase inhibitor, on renal injury
in hypercholesterolemia-induced hypertensive rats. Blood Press
14(2):120–125
62. Dubreuil M, Zhu Y, Zhang Y, Seeger JD, Lu N, Rho YH, Choi
HK (2015) Allopurinol initiation and all-cause mortality in the
general population. Ann Rheum Dis 74(7):1368–1372
63. Ma L, Hu J, Li J, Yang Y, Zhang L, Zou L, Gao R, Peng C, Wang
Y, Luo T, Xiang X, Qing H, Xiao X, Wu C, Wang Z, He JC, Li Q,
Yang S (2018) Bisphenol A promotes hyperuricemia via activat-
ing xanthine oxidase. FASEB J 32(2):1007–1016
64. Braga TT, Forni MF, Correa-Costa M, Ramos RN, Barbuto JA,
Branco P, Castoldi A, Hiyane MI, Davanso MR, Latz E, Franklin
BS, Kowaltowski AJ, Camara NO (2017) Soluble uric acid acti-
vates the NLRP3 inflammasome. Sci Rep 7:39884
65. He Y, Hara H, Nunez G (2016) Mechanism and regulation
of NLRP3 inflammasome activation. Trends Biochem Sci
41(12):1012–1021
66. Kim JJ, Jo EK (2013) NLRP3 inflammasome and host protection
against bacterial infection. J Korean Med Sci 28(10):1415–1423
Publisher’s Note Springer Nature remains neutral with regard to
jurisdictional claims in published maps and institutional affiliations.
... Isoorientin displayed weak XO inhibitory effect (IC50: 117.2 ± 13.5 µM) compared to positive control, quercetin (IC50: 7.7 ± 0.3 µM) [46]. An et al. also reported that isoorientin exerted 96% inhibition of XO enzyme at concentration of 667 µmol/L which is similar to allopurinol at 80 µmol/L concentration with 97% inhibitory ratio [47]. This study also proved that isoorientin showed weak inhibitory activity against XO which seems to be compatible with our results. ...
... It was reported that HU might increase the burden of the kidney and liver organs and cause tissues damages [49][50][51][52][53]. In the present study, the histopathological examination showed that the fruit and petiole extract markedly ameliorated renal and hepatic damage in HU rats, suggesting that A. longiloba extracts could significantly improve kidney and liver function. ...
Article
Full-text available
Hyperuricemia has become a significant public-health concern in recent years, and the available treatments have been reported to have an adverse side effect on patients. Alocasia longiloba has been used traditionally in Malaysia for treating gout, inflammation, and wounds. However, the plant has not been investigated for its effects on hyperuricemia. This study investigated the anti-hyperuricemic and anti-inflammatory effects of A. longiloba extracts in hyperuricemic rats induced by potassium oxonate (250 mg/kg body weight). Rats were given A. longiloba extracts or a standard drug for two-week, and blood and tissue samples were collected for analysis. Results show that A. longiloba extracts significantly reduced serum uric acid levels in hyperuricemic rats and inhibited xanthine oxidase (XOD) activity in the liver and kidney, which could be the mechanism underlying the urate-lowering effects. The extracts also significantly (p
... TLR4 is a receptor protein on the cell membrane, which can produce a signal to foreign stimuli, activate NF-kB, and produce inflammatory response [5]. Recent studies have shown that activation of NLRP3 and TLR4 contribute to the pathogenesis of UN [6,7]. These data indicate the potential of targeting the TLR4-NLRP3 signaling pathway as a novel therapeutic strategy. ...
Article
Full-text available
Background Uric acid nephropathy (UN) is a complication of hyperuricemia (HUA), which has a great impact on people's lives. Here, we evaluated the therapeutic potential of total flavonoids of Phellinus igniarius (TFPI) in vivo and studied the anti UN effect of TFPI in vitro. Methods Hyperuricemia was induced by intraperitoneal injection of potassium oxonate in ICR mice. After intervention with TFPI, we evaluated the levels of serum uric acid (UA) and creatinine (CR), and the contents of xanthine oxidase (XOD) and adenosine deaminase (ADA) in liver. To explore the effect and molecular mechanism of TFPI on UN, we treated HK-2 cells with monosodium urate (MSU) to study the effect of TFPI on apoptosis and inflammation. In addition, to explore the mechanism of TFPI on uric acid transport we evaluated the relationship between uric acid transporter ABCG2 and inflammatory signaling pathway TLR4-NLRP3. Results In the model mice, TFPI significantly decreased the levels of UA and Cr, which may be related to the inhibition of XOD enzyme activity. In HK-2 cells, the response of TFPI to MSU can effectively inhibit apoptosis and activation of TLR4-NLRP3 signaling pathway and promote the expression of ABCG2. Conclusions TFPI can significantly inhibit the release of inflammatory factors and promote the expression of ABCG2 by targeting TLR4 receptor and NLRP3 inflammasome. And targeted inhibition of XOD enzyme activity to reduce uric acid level and inhibit the development of UN.
... The global prevalence of HUA is 5-25%, another common metabolic disease after diabetes (31,32). As a key enzyme for the production of UA, xanthine oxidase (XOD) can directly catalyze the gradual oxidation of hypoxanthine and xanthine to form UA, and can also convert the rich protein in food into purine to finally form UA. Therefore, the inhibitors of XOD are widely used in the treatment of HUA (33)(34)(35)(36)(37)(38)(39). As a purine analog and hypoxanthine isomer, allopurinol (APL) is an important inhibitor of urate reduction (40)(41)(42) (46) found that by inhibiting the mRNA expressions, AST reduced UA synthesis and inhibited XOD and ADA enzyme activities, thereby alleviating HUA (46). ...
Article
Full-text available
Background As Gymnadenia R.Br. (Gym) has an obvious uric acid-lowering effect, but its specific bioactive substances and mechanism are still unclear. The key metabolites and pathways used by Gym to reduce uric acid (UA) were identify. Methods An optimized extraction process for urate-lowering active substances from Gym was firstly been carried out based on the xanthine oxidase (XOD) inhibition model in vitro; then, the Ultra-high-performance liquid chromatography and Q-Exactive mass spectrometry (UHPLC-QE-MS) based on non-targeted metabolomics analysis of Traditional Chinese Medicine were performed for comparison of Gym with ethanol concentration of 95% (low extraction rate but high XOD inhibition rate) and 75% (high extraction rate but low XOD inhibition rate), respectively; finally, the protective effect of ethanolic extract of Gym on zebrafish with Hyperuricemia (referred to as HUA zebrafish) was explored. Results We found that the inhibition rate of Gym extract with 95% ethanol concentration on XOD was 84.02%, and the extraction rate was 4.32%. Interestingly, when the other conditions were the same, the XOD inhibition rate of the Gym extract with 75% ethanol concentration was 76.84%, and the extraction rate was 14.68%. A total of 539 metabolites were identified, among them, 162 different metabolites were screened, of which 123 were up-regulated and 39 were down-regulated. Besides significantly reducing the contents of UA, BUN, CRE, ROS, MDA, and XOD activity in HUA zebrafish by Gym and acutely reduce the activity of SOD. Conclusion Along with the flavonoids, polyphenols, alkaloids, terpenoids, and phenylpropanoids, the ethanolic extract of Gym may be related to reduce the UA level of Gym.
Article
Objective Known for anti‐inflammatory and antioxidant properties, flavonoid has phytoestrogenic effects, but it is unclear whether its role in hyperuricemia and metabolic syndrome (MetS) differs by gender. Moreover, given the strong association between hyperuricemia and MetS, we aimed to explore whether flavonoid is a protective factor for hyperuricemia, independently of MetS, in different genders. Methods Data for 2007–2010 and 2017–2018 were obtained from the National Health and Nutrition Examination Survey (NHANES) and the Food and Nutrient Database for Dietary Studies (FNDDS). To assess the association among flavonoid, hyperuricemia, and MetS, multivariate logistic regression and subgroup analyses were conducted. Besides, to investigate whether the association between flavonoid and hyperuricemia was independent of MetS, multivariate logistic regression models were further conducted to explore the association between flavonoid and MetS among females with hyperuricemia and to investigate the association between flavonoid and hyperuricemia among females after excluding MetS. Result Among 5356 females, anthocyanin intake was inversely associated with the prevalence of hyperuricemia (Q4 vs. Q1: OR 0.49, 95% CI 0.31 to 0.76), and MetS (Q4 vs. Q1: OR 0.68, 95% CI 0.50 to 0.93). Furthermore, subgroup analyses showed the beneficial association between anthocyanin and hyperuricemia among females aged 40 to 59 years and menopausal. However, among 5104 males, no significant association was observed after adjustment for covariates (Q4 vs. Q1: OR 0.81, 95% CI 0.56 to 1.18). While in 372 females with hyperuricemia, no significant association was found between MetS and anthocyanin (Q4 vs. Q1: OR 0.88, 95% CI 0.31 to 2.49). Meanwhile, among 3335 females after excluding MetS, there was still a significant association between anthocyanin and a lower prevalence of hyperuricemia (Q4 vs. Q1: OR 0.38, 95% CI 0.17 to 0.85). Conclusion Dietary anthocyanin is associated with a lower prevalence of hyperuricemia independently of MetS among females. Foods rich in anthocyanin should be emphasized for females, especially those aged 40 to 59 years and menopausal, which may be of potential significance in the prevention of hyperuricemia.
Article
Full-text available
Gout represents a metabolic ailment resulting from the accumulation of monosodium urate crystals within joints, causing both inflammation and, harm to tissues. The primary contributor to gout’s emergence is an elevated presence of serum urate, which is under the regulation of kidney and, gut urate transporters. Mitigating this risk factor is crucial for averting gout’s onset. Several treatments rooted in TCM and related active compounds have demonstrated efficacy in managing gout, skillfully regulating serum uric acid (UA) levels and curbing inflammation’s progression. This analysis compiles key foundational research concerning the molecular signaling pathways and UA transporters linked to gout, under the regulation of TCM. The focus includes individual botanical drug, active compounds, and TCM formulations, which have been consolidated and examined in this overview. The primary keywords chosen were “gout, hyperuricemia, gouty arthritis, traditional Chinese medicine, Chinese botanical drug, medicinal botanical drug, and natural plant”. Various relevant literature published within the last 5 years were gathered from electronic databases, including PubMed, Web of Science, CNKI, and others. The findings revealed that TCM has the capacity to modulate various signaling pathways, including MAPK, NF-κB, PI3K/Akt, NLRP3 and JAK/STAT. Additionally, it impacts UA transporters like URAT1, GLUT9, ABCG2, as well as OATs and OCTs, thereby contributing to gout treatment. TCM helps maintain a balanced inflammatory interaction and facilitates UA excretion. This study enhances our understanding of TCM’s anti-gout mechanisms and introduces novel perspectives for establishing the clinical significance and future prospects of TCM-based gout treatment.
Article
Full-text available
Hyperuricemia (HUA) is a metabolic disease caused by excessive uric acid in patients. Flavonoids have attracted extensive attention due to their safety, effectiveness, and little side effects in the treatment of HUA and inhibition of xanthine oxidase (XO) activity. In this paper, the studies on the treatment of HUA and the inhibition of XO activity by flavonoids (including flavones, flavonols, flavanones, flavanols, isoflavonoids, anthocyanins, and chalcones) were comprehensively reviewed from the aspects of structure–activity relationship and animal experiments. Results showed that hydrogen bonds and hydrophobic interactions were the two most important forces involved in the interaction between flavonoids and XO. Flavonoids could combine with the hydrophobic cavity of XO to cause changes in XO structure and influence the action of enzyme and substrate, thereby decreasing the catalytic activity of XO. Moreover, flavonoids undergo a series of metabolic reactions in the organism, which plays a role in alleviating HUA by affecting the expression of related genes and proteins (e.g., glucose transporter 9, urate transporter 1, and organic anion transporter 1) in the kidneys, liver, and intestinal tract of the organism and changing the internal environment in organs. In addition, this paper provides new perspectives on the shortcomings of the current research, to promote the in‐depth development and application of flavonoids.
Article
Full-text available
Cardiovascular diseases (CVDs) are a leading cause of global mortality and have a high incidence rate worldwide. The function of inflammasomes in CVDs has received a lot of attention recently, and the nucleotide-binding domain and leucine-rich repeat protein 3 (NLRP3) inflammasome may be a new target for the prevention and treatment of CVDs. Flavonoids, which are found in food and plant extracts, inhibited inflammation in CVDs by regulating the NLRP3 inflammasome. CB-Dock was used to investigate whether 34 flavonoids from natural products acted on NLRP3 inflammasome. In brief, the PDB format of NLRP3 was selected as a protein file, and 34 flavonoids in SDF format were selected as the ligand file, and then input to CB-Dock for molecular docking. The docking results showed that epigallocatechin-3-gallate (EGCG), amentoflavone, baicalin, scutellarin, vitexin, silibinin, and puerarin had good binding affinities to NLRP3, which could be used as NLRP3 inhibitors, and aid in the discovery of lead compounds for the design and development of CVDs.
Article
Full-text available
Hesperetin is a natural flavonoid with many biological activities. In view of hyperuricemia treatment, the effects of hesperetin in vivo and in vitro, and the underlying mechanisms, were explored. Hyperuricemia models induced by yeast extract (YE) or potassium oxonate (PO) in mice were created, as were models based on hypoxanthine and xanthine oxidase (XOD) in L-O2 cells and sodium urate in HEK293T cells. Serum level of uric acid (UA), creatinine (CRE), and urea nitrogen (BUN) were reduced significantly after hesperetin treatment in vivo. Hesperetin provided hepatoprotective effects and inhibited xanthine oxidase activity markedly, altered the level of malondialdehyde (MDA), glutathione peroxidase (GSH-PX) and catalase (CAT), downregulated the XOD protein expression, toll-like receptor (TLR)4, nucleotide binding oligomerization domain-like receptor family pyrin domain-containing 3 (NLRP3) inflammasome, interleukin-18 (IL-18), upregulated forkhead box O3a (FOXO3a), manganese superoxide dismutase (MnSOD) in a uric acid-synthesis model in mice. Protein expression of organic anion transporter 1 (OAT1), OAT3, organic cationic transporter 1 (OCT1), and OCT2 was upregulated by hesperetin intervention in a uric acid excretion model in mice. Our results proposal that hesperetin exerts a uric acid-lowering effect through inhibiting xanthine oxidase activity and protein expression, intervening in the TLR4-NLRP3 inflammasome signaling pathway, and up-regulating expression of FOXO3a, MnSOD, OAT1, OAT3, OCT1, and OCT2 proteins. Thus, hesperetin could be a promising therapeutic agent against hyperuricemia.
Article
Hyperuricemia is a metabolic disease caused by the accumulation of uric acid in the body. Allopurinol, benzbromarone, and febuxostat, which are available in the market, have reduced the circulating urate levels; however, they exhibit serious side effects. Therefore, it is reasonable to develop a new active antihyperuricemia drug with few side effects. With the deepening of research, numerous kinds of literature have shown that natural active substances are effective in the treatment of hyperuricemia with a variety of sources and few side effects, which have become the focus of research in recent years. This review focuses on natural active substances with uric-acid-reducing activity and discusses their pharmacological effects. More specifically, the bioactive compounds of natural active substances are divided into five categories: natural extracts, monomer compounds extracted from plants, natural protease hydrolysates, peptides, and probiotic bacteria. In addition, the mechanisms by which these bioactive compounds exhibit hypouricemic effects can be divided into four classes: inhibition of key enzyme activities, promotion of uric acid excretion and inhibition of reabsorption in the kidney, promotion of decomposing uric acid precursors, and promotion of decomposing uric acid. Overall, this current and comprehensive review examines the role of natural active substances in the treatment of hyperuricemia.
Article
Full-text available
Lysosomal cathepsin B (CTSB) has been proposed to play a role in the induction of acute inflammation. We hypothesised that the presence of active CTSB in the cytosol is crucial for NLRP3-inflammasome assembly and, consequently, for mature IL-1β generation after mycobacterial infection in vitro. Elevated levels of CTSB was observed in the lungs of mice and rabbits following infection with Mycobacterium tuberculosis (Mtb) H37Rv as well as in plasma from acute tuberculosis patients. H37Rv-infected murine bone marrow-derived macrophages (BMDMs) displayed both lysosomal leakage, with release of CTSB into the cytosol, as well as increased levels of mature IL-1β. These responses were diminished in BMDM infected with a mutant H37Rv deficient in ESAT-6 expression. Pharmacological inhibition of cathepsin activity with CA074-Me resulted in a substantial reduction of both mature IL-1β production and caspase-1 activation in infected macrophages. Moreover, cathepsin inhibition abolished the interaction between NLRP3 and ASC, measured by immunofluorescence imaging in H37Rv-infected macrophages, demonstrating a critical role of the enzyme in NLRP3-inflammasome activation. These observations suggest that during Mtb infection, lysosomal release of activated CTSB and possibly other cathepsins inhibitable by CA07-Me is critical for the induction of inflammasome-mediated IL-1β processing by regulating NLRP3-inflammasome assembly in the cytosol.
Article
Full-text available
BACKGROUND AND OBJECTIVES: Recent studies have shown that hyperuricemia is commonly associated with dyslipidemia, cardiovascular diseases, hypertension and metabolic syndrome. Elevated serum uric acid has been demonstrated to be associated with obesity in the adult population in many countries; however, there is still a lack of evidence for the Bangladeshi population. The aims of this study were to evaluate the prevalence of hyperuricemia and determine the relationship between serum uric acid (SUA) and obesity among the Bangladeshi adults. METHODS: In this cross-sectional study, blood samples were collected from 260 adults (142 males and 118 females) and analyzed for SUA and lipid profile. All participants were categorized as underweight (n = 11), normal (n = 66), overweight (n = 120) and obese (n = 63) according to the body mass index (BMI) scale for the Asian population. Based on SUA concentration the participants were stratified into four quartiles (Q1: < 232 μmol/L, Q2: 232-291 μmol/L, Q3: 292-345 μmol/L and Q4: > 345 μmol/L). RESULTS: The mean age and BMI of the participants were 32.5 ± 13.3 years and 24.9 ± 3.8 kg/m2, respectively. The average level of SUA was 294 ± 90 μmol/L with a significant difference between males and females (p < 0.001). Overall, the estimated prevalence of hyperuricemia was 9.3% with 8.4% in male and 10.2% in female participants. There were significant increases in the prevalence of obesity (17.4%, 22.2%, 28.6% and 31.8%, respectively, p < 0.01 for trend) across the SUA quartiles. A multiple logistic regression analysis revealed that SUA quartiles were independently associated with the presence of obesity (p < 0.01). CONCLUSION: Present study indicates a significant positive relationship between SUA and obesity among the Bangladeshi adults. Therefore, routine measurement of SUA is recommended in obese individuals to prevent hyperuricemia and its related complications.
Article
Full-text available
We report the case of a 60-year old patient with a 1-month history of hyperuricemia treated with allopurinol. The patient presented to the Department of Dermatology with acute rash on the face and the lower limbs associated with fever, arthralgias and myalgias. Clinical examination showed symmetric macular erythema on the face at the level of the cheeks with discreet edema (A), erythematous plaques at the level of both legs with healthy skin areas extending progressively from the bottom-up. Lesions were very itchy with burning sensation (B). The examination of the oral cavity showed very painful erosive lesions at the level of the internal face of the cheeks. Lymph nodes were free. Paraclinical tests showed leukocyte counts at 20000, elevated transaminases > 100 IU, eosinophil counts at 1500, HHV6 serology was negative. The diagnosis of DRESS syndrome was retained. The patient underwent corticosteroid therapy at a dose of 1 mg/kg/day associated with symptomatic treatment. Treatment evolution was marked by spectacular improvement after 06 days (C and D) with regression of skin lesions and gradual normalization of laboratory results.
Article
Full-text available
Background The xanthine oxidase inhibitor allopurinol improves endothelial function in different populations, including patients with chronic heart failure (CHF). Its effect on arterial stiffness parameters is less clear. We investigated the effect of short-term low-dose allopurinol therapy on arterial stiffness in Saudi patients with stable mild-moderate CHF. Methods A prospective, randomized, double-blind, placebo-controlled study was performed on 73 patients with mild-moderate CHF. In all, 36 patients were randomized to allopurinol 300 mg daily for 3 months, while 37 patients were randomized to placebo. Arterial stiffness parameters, aortic pulse wave velocity (Ao-PWV) and heart rate corrected augmentation index (c-AIx), were assessed before and after treatment along with serum uric acid. Results A total of 66 patients completed the study. Both groups were matched for age, sex, severity of heart failure, and arterial stiffness. Compared with placebo, allopurinol recipients had a significant fall in uric acid concentration from 6.31 ± 1.4 (SD) mg/dL to 3.81 ± 1.2 (P < .001). Despite that, there was no significant change in arterial stiffness parameters between allopurinol and placebo groups. Post-treatment Ao-PWV was 9.79 ± 2.6 m/s in the allopurinol group and 10.07 ± 3.4 m/s in the placebo group, P = .723. Post-treatment c-AIx was 24.0% ± 9.1% and 22.0% ± 9.9%, respectively, P = .403. Conclusions We have shown that allopurinol significantly reduced uric acid concentration in Saudi patients with CHF but was not associated with a change in arterial stiffness. Our cohort of patients had worse arterial stiffness values at baseline, which might make them more resistant to change using our study regimen. The study has been registered with the International Standard Randomized Controlled Trial Number registry with an identifier number of ISRCTN58980230.
Article
Full-text available
A mangiferin aglycon derivative J99745 has been identified as a potent xanthine oxidase (XOD) inhibitor by previous in vitro study. This study aimed to evaluate the hypouricemic effects of J99745 in experimental hyperuricemia mice, and explore the underlying mechanisms. Mice were orally administered 600 mg/kg xanthine once daily for 7 days and intraperitoneally injected 250 mg/kg oxonic acid on the 7th day to induce hyperuricemia. Meanwhile, J99745 (3, 10, and 30 mg/kg), allopurinol (20 mg/kg) or benzbromarone (20 mg/kg) were orally administered to mice for 7 days. On the 7th day, uric acid and creatinine in serum and urine, blood urea nitrogen (BUN), malondialdehyde (MDA) content and XOD activities in serum and liver were determined. Morphological changes in kidney were observed using hematoxylin and eosin (H&E) staining. Hepatic XOD, renal urate transporter 1 (URAT1), glucose transporter type 9 (GLUT9), organic anion transporter 1 (OAT1) and ATP-binding cassette transporter G2 (ABCG2) were detected by Western blot and real time polymerase chain reaction (PCR). The results showed that J99745 at doses of 10 and 30 mg/kg significantly reduced serum urate, and enhanced fractional excretion of uric acid (FEUA). H&E staining confirmed that J99745 provided greater nephroprotective effects than allopurinol and benzbromarone. Moreover, serum and hepatic XOD activities and renal URAT1 expression declined in J99745-treated hyperuricemia mice. In consistence with the ability to inhibit XOD, J99745 lowered serum MDA content in hyperuricemia mice. Our results suggest that J99745 exerts urate-lowering effect by inhibiting XOD activity and URAT1 expression, thus representing a promising candidate as an anti-hyperuricemia agent.
Article
Full-text available
Background In addition to the kidney, the intestine is one of the most important organs involved in uric acid excretion. However, the mechanism of urate excretion in the intestine remains unclear. Therefore, the relationship between soluble uric acid and the gut excretion in human intestinal cells was explored. The relevant signaling molecules were then also examined. MethodsHT-29 and Caco-2 cell lines were stimulated with soluble uric acid. Western blotting and qRT-PCR were used to measure protein and mRNA levels. Subcellular fractionation methods and immunofluorescence were used to quantify the proteins in different subcellular compartments. Flow cytometry experiments examined the function of ATP-binding cassette transporter, subfamily G, member 2 (ABCG2). Small interfering RNA transfection was used to assess the interaction between ABCG2 and PDZ domain-containing 1 (PDZK1). ResultsSoluble uric acid increased the expression of PDZK1 and ABCG2. The stimulation of soluble uric acid also facilitated the translocation of ABCG2 from the intracellular compartment to the plasma membrane and increased its transport activity. Moreover, the upregulation of PDZK1 and ABCG2 by soluble uric acid was partially decreased by either TLR4-NLRP3 inflammasome inhibitors or PI3K/Akt signaling inhibitors. Furthermore, PDZK1 knockdown significantly inhibited the expression and transport activity of ABCG2 regardless of the activation by soluble uric acid, demonstrating a pivotal role for PDZK1 in the regulation of ABCG2. Conclusions These findings suggest that urate upregulates the expression of PDZK1 and ABCG2 for excretion in intestinal cells via activating the TLR4-NLRP3 inflammasome and PI3K/Akt signaling pathway.
Article
Lemon is a healthy fruit with high medicinal value. This study found that lemon water soluble extract (LET) can reduce uric acid levels in mice with potassium oxonate induced hyperuricemia. Histopathological analysis suggested that LET caused little damage to the kidneys of mice. It affected mABCG2 and mGLUT9 mRNA expression only in hyperuricemic mice, but not in healthy mice. Our further results show that potassium citrate, rather than citric acid, is the main ingredient in LET with a hypouricemic effect. This study also indicates that lemon does have unique medicinal value for the treatment of hyperuricemia, and that potassium citrate has the potential to be developed as a drug for hyperuricemia. Lowering uric acid through LET and potassium citrate may directly promote the degradation of excessive uric acid in patients with hyperuricemia.
Article
The abnormal metabolism of uric acid results in many disease such as chronic kidney disease, hyperuricemia, nephrolithiasis, gout, hypertension, vascular disease and so on. Serum uric acid levels are maintained by the balance between production and elimination. There are many factors that maintain the balance of serum uric acid. One of them is transporters which are responsible for the debouchment of uric acid within blood. The transport and excretion of uric acid is a complicated procedure which is related with various transporters such as OAT1, OAT3, OAT4, URAT1, GLUT9, BCRP, MRP4, NPT1, NTP4, and so on. In recent years, a large number of genome-wide association studies have shown that the single nucleotide polymorphisms of uric acid transporters were closely related to serum uric acid level. What’s more, some mutations on these gene locus may also break the balance of serum uric acid. Here, the polymorphisms of uric acid transporters closely related with the serum uric acid balance were reviewed and discussed because of their important significance in clinical therapy for a precision medicine. The mechanism of metabolic diseases with gene variation may provide new strategy for the design and development of innovative drug to treat diseases with uric acid metabolic disturbance.
Article
Uric acid is the final product of purine metabolism. Hyperuricemia is defined as a condition where the level of uric acid exceeds the normal range. The most well-known disease induced by hyperuricemia is gout. However, many studies have reported that hyperuricemia also plays important roles in cardiac-kidney-vascular system diseases and metabolic syndrome. Although hyperuricemia has been known for a long time, its pathophysiology remains poorly understood. In this review, we highlight studies on advanced pathological mechanisms for injuries induced by hyperuricemia, summarize epidemiological studies on hyperuricemia and its associated diseases, and take a brief look at hyperuricemia prevention.
Article
Hyperuricemia is not only a risk factor for gout but also an independent determinant of hypertension, diabetes, and chronic kidney diseases. Although the incidence of gout in Korean adults is increasing, epidemiologic studies on hyperuricemia in the general Korean population are limited. Thus, this study aimed at evaluating the prevalence of hyperuricemia and its associated factors among non-institutionalized Korean adults. The present study included 5548 participants (2403 men and 3145 women) aged ≥ 19 years from The Korea National Health and Nutrition Examination Survey. Based on the new 2016 census data, the age-standardized prevalence and mean uric acid level were calculated using the chi-square test and t test, respectively. A multivariate logistic regression analysis was performed to evaluate the risk factors associated with hyperuricemia. The age-standardized prevalence of hyperuricemia and mean uric acid level in the general Korean population was 11.4% (17.0% in men and 5.9% in women) and 5.1 mg/dL (5.83 mg/dL in men and 4.36 mg/dL in women), respectively. The prevalence of hyperuricemia was high in young Korean adults, and a U-shaped association was observed between hyperuricemia and age. While obesity, metabolic syndrome, renal impairment, and low-grade inflammation were positively associated with hyperuricemia in both sexes, alcohol consumption, education, and current smoking status had a positive association with hyperuricemia only in women. Hyperuricemia is prevalent in the young population in Korea, and special efforts are necessary to reduce the potential harmful effects of hyperuricemia on the health of adults, particularly the younger-generation adults, in Korea.