You are on page 1of 10

pubs.acs.

org/JAFC Article

Role of Thioredoxin-Interacting Protein in Diabetic Fatty Kidney


Induced by Advanced Glycation End-Products
Hong Sun,*,⊥ Juan Chen,⊥ Lili Sun, Bimin Shi, and Jianzhong Li*

Cite This: https://doi.org/10.1021/acs.jafc.1c03559 Read Online

ACCESS Metrics & More Article Recommendations


See https://pubs.acs.org/sharingguidelines for options on how to legitimately share published articles.

ABSTRACT: Advanced glycation end-products (AGEs) have been identified as the etiological factors associated with the fatty
Downloaded via UNIV ESTADUAL PAULISTA on October 9, 2021 at 20:18:42 (UTC).

kidney. Thioredoxin-interacting protein (TXNIP) might be a mediator involved in AGE-induced fatty kidney. This study focused on
investigating how TXNIP affected the AGE-mediated renal lipid deposition. In an in vivo experiment, the db/db mice injected with
the lentiviral vector encoding shRNA targeting TXNIP were given the AIN-76 basal or the high-AGE diet. TXNIP-targeting siRNA-
transfected human renal proximal tubular epithelial (HK-2) cells were exposed to AGE-BSA in a study in vitro. The results showed
that the silencing of TXNIP reduced tubular lipid droplets and intracellular cholesterol content, as well as upregulated Insig-1 and
downregulated HMGCoAR, LDLr, nSREBP-2, and SCAP in the kidneys of the db/db mice, the high-AGE-diet-fed db/db mice, and
AGE-BSA-treated HK-2 cells. Furthermore, AGE-BSA enhanced SCAP−SREBP-2 complex formation while promoting their
transportation to the Golgi apparatus. However, these could be inhibited by TXNIP silencing in the HK-2 cells. The above findings
indicated that TXNIP knockdown mitigated the accumulation of renal tubular lipids in diabetes through the regulation of SCAP,
thereby inhibiting the SCAP−SREBP-2 signaling pathway, resulting in reduced cholesterol uptake and synthesis. Therefore, TXNIP
might be a potential therapeutic target to treat a diabetic fatty kidney.
KEYWORDS: type 2 diabetes mellitus, fatty kidney, advanced glycation end-products, thioredoxin-interacting protein

■ INTRODUCTION
The latest findings presented by the International Diabetes
that renal tubular lesions occur earlier than glomerular lesions,
and the severity of the renal tubular injury has become an
Federation (IDF) show that the incidence of diabetes mellitus important indicator of the progression and prognostic outcome
(DM) has been increasing rapidly worldwide, and the number of kidney function of DM patients.6,7 Among the pathogenic
of diabetes patients is expected to exceed 435 million by 2030.1 factors associated with DM, an excess of advanced glycation
DM and its associated organ damage seriously affect people’s end-products (AGEs) have been recognized as the critical
health and quality of life. In recent years, the diabetes-related driving factor for renal tubular injury in patients with T2DM.
AGEs can be produced during the nonenzymatic reaction of
fatty kidney has been reported increasingly frequently. The
protein amino groups with reducing sugars, which have key
concept of the fatty kidney was first proposed as early as the
functions in the pathogenic mechanisms of DM as well as the
1900s. However, it was not until John Moorhead proposed the
related complications. Our previous studies have shown that
hypothesis of renal lipid toxicity in 1982 that fatty kidney
excessive AGEs can disrupt intracellular cholesterol feedback
began to attract the attention of experts.2 Lipometabolic
regulation, thereby leading to the accumulation of intracellular
disturbance induces lipid ectopic deposits in the kidney and
lipids within the renal proximal tubular epithelial cells (HK-2
caused the development of fatty kidney, which is a common
cells) and promoting the development of a fatty kidney.8,9
phenomenon in type 2 diabetes mellitus (T2DM). Evidence
Sterol regulatory element-binding protein (SREBP)-cleavage
suggests that the renal lipid contents among T2DM patients
activating protein (SCAP) modulates intracellular cholesterol
detected through magnetic resonance imaging (MRI) remark-
feedback. SCAP is not only an intracellular cholesterol sensor
ably increased relative to those among non-T2DM patients.3,4
but also SREBP-2’s molecular chaperone. Upon the demand
Moreover, histological staining indicated abnormal deposition
for cholesterol by cells, SCAP separates with insulin-induced
of lipid droplets in renal glomeruli, mesangia, interstitium, and
gene-1 (Insig-1), the endoplasmic reticulum (ER) protein,
especially, renal tubules in T2DM patients,2,5 which not only
while delivering SREBP-2 within ER into Golgi apparatus and
represents the formation of fatty kidney but is also associated
with impairment of renal morphology and function. Therefore,
it is urgently important to investigate the pathogenic Received: June 15, 2021
mechanism by which fatty kidney develops in association Revised: September 22, 2021
with T2DM. Accepted: September 23, 2021
Previously, glomerular injury has a central effect on the
pathogenic mechanism of diabetic kidney disease (DKD).
Nonetheless, an increasing number of studies have reported

© XXXX American Chemical Society https://doi.org/10.1021/acs.jafc.1c03559


A J. Agric. Food Chem. XXXX, XXX, XXX−XXX
Journal of Agricultural and Food Chemistry pubs.acs.org/JAFC Article

activating it through proteolytic cleavage. After cleavage, the by subjecting the AIN-76 basal diet to 10 min of heat exposure at 90
N-terminal in SREBP-2 (nSREBP-2) experiences nuclear °C for generating the diet-mediated AGEs.22 Then, all animals were
translocation to activate transcriptional factors including 3- classified into six groups, with five animals each. Group 1: control db/
hydroxy-3-methylglutaryl coenzyme A reductase (HMGCoAR, m mice were exposed to the AIN-76 basal diet (db/m); Group 2: db/
db mice were exposed to the AIN-76 basal diet (db/db); Group 3:
a key enzyme involved in cholesterol production) as well as db/db mice were exposed to sh-NC and the AIN-76 basal diet (db/
low-density lipoprotein receptor (LDLr, a cholesterol uptake db + sh-NC); Group 4: db/db mice injected with sh-TXNIP and fed
channel), leading to increased synthesis and uptake of an AIN-76 basal diet (db/db + sh-TXNIP); Group 5: db/db mice fed
cholesterol. By contrast, Insig-1−SCAP−SREBP complexes a high-AGE diet (db/db + AGEs); and Group 5: db/db mice were
are reserved within ER once there is enough cholesterol within exposed to sh-TXNIP and fed high-AGE diet (db/db + AGEs + sh-
cells, which prevents cholesterol overloading in cells under TXNIP). The animals were raised in separate metabolic cages to
physiological conditions.10,11 However, the specific mechanism collect the 24 h urine. When the experiment was completed, the blood
underlying AGE-induced abnormal cholesterol feedback was sampled to conduct biochemical tests, and renal tissues were
regulation is still elusive and so further research is warranted. employed to assess the histology.
Biochemical Tests. Each animal was killed upon the completion
Thioredoxin-interacting protein (TXNIP), referred to as
of the experiment. Blood was sampled in the right ventricle to carry
thioredoxin-binding protein-2 (TBP-2) or vitamin D3 out biochemical tests, like serum creatinine (SCR), fasting blood
upregulated protein 1 (VDUP1), is closely related to metabolic glucose (FBG), total cholesterol (TC), total triglycerides (TG), and
disorders like diabetes, fatty liver, obesity, and atheroscle- blood urea nitrogen (BUN) by the use of automatic analyzers
rosis.12−14 Recently, some clinical studies have shown that the (Hitachi). Serum AGEs and markers related to renal tubular injuries,
level of TXNIP gene in the peripheral blood cells of T2DM such as urinary neutrophil gelatinase-associated lipocalin (u-NGAL)
patients is positively correlated with serum AGEs levels,15 and and β2-microglobulin (β2-MG), were detected by ELISA (CUSA-
another animal study has shown that AGEs could upregulate BIO, Wuhan, China). In addition, Coomassie Brilliant Blue protein
the expression of TXNIP in the kidneys of BALB/c mice,16 assay (Jiancheng Bioengineering Institute, Nanjing, Jiangsu) was
indicating AGEs might be stimulators that regulate the adopted for measuring the 24 h urine proteins.
Renal Morphology. Renal cortical tissue was prepared into
expression of TXNIP. Additionally, Chang Hyun Byon et al. paraffin-embedded sections in sequence. Then, each cross section
reported that knocking out the TXNIP gene reduces lipid (measuring 3 μm) was put in the gelatin-coated slides and subjected
deposition in the arterial intima of ApoE−/− mice, thereby to hematoxylin-eosin (HE), periodic acid Schiff (PAS), and Masson
alleviating atherosclerosis.17 Other researchers found that and immuohistochemical (IHC) staining.
TXNIP deletion ameliorates HFD-induced hepatic steatosis Cell Culture. This study acquired HK-2 cells at the American
in C57Bl/6J wild-type mice.18 Furthermore, Wang W et al. Type Culture Collection (Manassas, VA), cultivated them within the
demonstrated that reducing the expression of TXNIP can RPMI-1640 medium HyClone (Logan, Utah) that contained 10%
lower SREBP-2-mediated liver cholesterol synthesis, resulting fetal bovine serum (FBS, HyClone; GE Healthcare Life Sciences,
in the alleviation of fatty liver disease in type 1 diabetic rats.19 Logan, UT), 1% streptomycin/penicillin (Merck KGaA; Sigma-
Aldrich), and 2 mM L-glutamine solution, and incubated them under
Hence, we suspect that TXNIP might be involved in the 37 °C, 5% CO2 and 95% humidity conditions for a period of 24 h.
abnormal feedback regulation of cholesterol, which may be Each experiment was carried out within the serum-free RPMI-1640
attributed to tubular foam cell formation. Based on the findings medium supplemented with 0.2% bovine serum albumin (BSA;
described above, this work focused on investigating how Sigma-Aldrich; Merck KGaA). AGE-BSA was purchased from Abcam
TXNIP affected the AGE-mediated renal lipid accumulation, (Cambridge, U.K.). We cultured HK-2 cells within the 200 μg/ml
especially cholesterol synthesis and uptake, in diabetic mice fed AGE-BSA-containing medium or the experimental medium for 48 h.
high-AGE diets and subjected to TXNIP silencing. We also Small Interfering (si) RNA Transfection. This study cultured
attempted to elucidate the exact mechanism of TXNIP in HK-2 cells (5 × 105 cells/well) into six-well plates and transiently
regulating lipid metabolism within the AGE-mediated human transfected them with TXNIP siRNA (sense: 5′-CTCCCUG-
CUAUAUGGAUGUtt-3′; antisense: 5′-ACAUCCAUAUAGCAGG-
HK-2 cells at the molecular level.


GAGtt-3′) or empty vector siRNA (Enhancer Biotechnology Co.,
Ltd., Nanjing, China) with Lipofectamine 3000 (Invitrogen; Thermo
MATERIALS AND METHODS Fisher Scientific, Inc., Waltham, MA) in line with specific protocols,
Animal Experimental Design. This study acquired male db/m which was performed as previously described.23
mice together with the C57BL/KsJ db/db mice at the National Model Observation of Lipid Accumulation. Renal lipid deposition
Animal Center of Nanjing University (Nanjing, China). The study within HK-2 cells and the db/db mice was evaluated by Oil Red O
protocols gained approval from the Ethics Committee of Soochow staining. Briefly, 4% paraformaldehyde was used to fix samples,
University in line with the Declaration of Helsinki and in compliance followed by 30 min of Oil Red O staining. Subsequently, hematoxylin
with the ARRIVE guidelines. Animals were raised within the was adopted to counterstain samples for a period of 5 min. A light
polypropylene cages under the following conditions, light/dark microscope (Carl Zeiss, Hertfordshire, U.K.) was employed for the
cycle of 12/12 h, room temperature of 22 ± 2 °C, and relative result examination.
humidity (RH) of 60 ± 5%. Each mouse was adaptively fed for 2 Quantification of Intracellular Cholesterol. Free cholesterol
weeks, then alcohol was used to wipe their tails for tail vein expansion (FC) and intracellular TC levels were quantitatively measured in vivo
for subsequent injection. Later, each animal was given a slow injection and in vitro by enzymatic assays (Applygen Technologies Inc., Beijing,
of 100 μl of a lentiviral vector encoding shRNA targeting TXNIP China). In brief, after sample collection, lipids were extracted with 1
(Genepharma, Shanghai, China) at 1 × 107 TU/ml by the 0.5 ml mL of chloroform/methanol (2:1). Afterward, the samples were
syringe (injection velocity, 0.2 μL/min) via tail vein, and the sequence sonicated and centrifuged to obtain the lipid phase. Thereafter, we
of sh-TXNIP was established as GTCTCTGCTCGAATTGACA (5′- treated samples according to specific instructions. The standard curve
3′). Equal volumes of the negative plasmid (sh-NC) or saline were was utilized to analyze FC and TC contents in each sample.
injected into the control groups. To ensure that the downregulation of Cholesterol ester (CE) level was measured by subtracting TC by FC.
TXNIP was induced, injection was administered at weeks 1, 3, 5, and Protein Extraction and Western Blot (WB) Analysis. The total
7.20,21 One group of 8-week-old db/db mice was given the AIN-76 cellular and nuclear proteins were collected, followed by denaturation
basal diet (Xietong Biology Co., Ltd., Nanjing, China), and another and 6%−15% SDS-PAGE, which was performed as previously
group of these mice was given a high-AGE diet, which was prepared described.24 Later, proteins were transferred onto poly(vinylidene

B https://doi.org/10.1021/acs.jafc.1c03559
J. Agric. Food Chem. XXXX, XXX, XXX−XXX
Journal of Agricultural and Food Chemistry pubs.acs.org/JAFC Article

Table 1. Biochemical Characteristics of the Micea


parameters db/m db/db db/db + sh-NC db/db + sh-TXNIP db/db + AGEs db/db + AGEs + sh-TXNIP
FBG (mmol/L) 6.33 ± 0.25 18.83 ± 1.48b 17.35 ± 0.67 11.78 ± 1.40c 29.18 ± 1.36c 22.43 ± 1.83d
AGEs (ng/mL) 1.22 ± 0.14 3.97 ± 0.19b 3.56 ± 0.21 3.57 ± 0.26 5.94 ± 0.25c 5.54 ± 0.42
TG (mmol/L) 1.22 ± 0.09 2.43 ± 0.18b 2.57 ± 0.23 2.37 ± 0.16 2.66 ± 0.16 2.60 ± 0.26
TC (mmol/L) 2.27 ± 0.04 3.46 ± 0.16b 3.29 ± 0.18 3.02 ± 0.21 3.36 ± 0.16 3.30 ± 0.13
body weight (g) 21.55 ± 0.52 50.95 ± 0.36b 49.78 ± 0.62 50.88 ± 2.22 52.85 ± 1.57 49.78 ± 1.11
food intake (g/day) 5.48 ± 0.62 6.35 ± 0.96 5.92 ± 1.14 6.58 ± 1.32 6.86 ± 1.42 6.04 ± 1.10
water intake (mL/day) 5.60 ± 0.78 22.53 ± 3.21b 20.66 ± 2.98 14.52 ± 2.12c 28.00 ± 3.42c 16.04 ± 2.78d
a
The levels of FBG, AGEs, TG, TC, the body weight, the food intake, and the water intake of the mice. Values are expressed as the mean ± S.E.M.
of five mice in each group. bP < 0.01 versus the db/m group. cP < 0.01 versus the db/db group. dP < 0.01 versus the AGEs + db/db group.

fluoride) (PVDF) membranes (GE Healthcare, Buckinghamshire, of FBG, compared with the db/db mice or db/db mice
U.K.), followed by 1 h of blocking using 5% BSA within Tris-buffered exposed to high-AGEs diet, while AGEs, TG, and TC exhibited
saline supplemented with 0.05% Tween 20 (TBST) under ambient no changes. In addition, compared with that in the db/m mice,
temperature. After washing, we incubated the blots with 5% BSA
polydipsia was observed in the db/db mice, especially in the
within TBST that contained diluted antibodies against TXNIP, SCAP,
SREBP-2, LDLr, HMGCoAR, Insig-1, Lamin A, and GAPDH db/db mice exposed to high-AGE diets, and silencing of
(Abcam, Cambridge, U.K.) under 4 °C overnight. After rinsing with TXNIP decreased the water intake in the db/db mice exposed
TBST thrice, the secondary antibody diluted at 1:5000 with TBST to basal diet and those exposed to high-AGE diet. However,
that contained 1% BSA (Santa Cruz Biotechnology) was used to the difference in food intake was not significant among mice.
further incubate the membranes under ambient temperature for 1 h, Silencing of TXNIP Reduced Renal Tubular Lipid
followed by rinsing by TBST thrice again. Thereafter, the Accumulation In Vivo and In Vitro. For exploring the
chemiluminescence reaction (Pierce, Rockford, IL) was conducted involvement of TXNIP in regulating in vivo abnormal lipid
and blots were exposed to X-ray film for a suitable period of time to deposition within diabetic kidneys, this study analyzed renal
detect bands. Later, LabWorks software (UVP Laboratory Products,
Upland, CA) was utilized for densitometrical detection and evaluation
lipid droplet deposition in diabetic mice transfected with sh-
of bands, and the bands were normalized with the Lamin A or TXNIP or a control plasmid. This work first verified TXNIP
GAPDH bands. depletion within the kidneys of the db/db mice through IHC
Confocal Microscopy. The human SCAP cDNA was ligated at staining and Western blot analysis (Figure 1A,B). Then, Oil
BstE-XbaI sites in pEGFP-C1 vector (Genechem Co. Ltd., Shanghai, Red O staining was conducted. Strong positive areas were
China) to prepare the green fluorescent protein (GFP)-SCAP found within renal tubules from the db/db mice, and nearly no
expression construct, which was performed as previously described.8 stain was observed in the db/m mice (Figure 2A). Intracellular
Effectene Transfection Reagent (Invitrogen, Paisley, U.K.) was cholesterol level was quantified, which verified the increase in
utilized to transfect pEGFP-SCAP into cells in accordance with the level of renal cholesterol ester deposition in the db/db
specific instructions. Subsequently, we placed cells onto chamber
slides to incubate them within the growth medium for a period of 48 mice (Figure 2B). Moreover, significantly increased intra-
h, followed by 30 min of fixation with 5% formalin solution, 15 min of cellular cholesterol contents and Oil Red O staining were
permeabilization using 0.25% Triton X-100, and 2 h of staining using observed among the db/db mice fed with high-AGE diet
Golgin-97 antibodies (Abcam, Cambridge, U.K.) under ambient compared with those fed with basal diet. However, silencing of
temperature. Later, cells were rinsed and counterstained with the TXNIP reduced intracellular cholesterol and tubular lipid
secondary fluorescent antibody for an additional one hour. The droplet contents in the db/db mice exposed to high-AGE and
resulting cells were examined by confocal microscopy using Zeiss basal diets (Figure 2A,B).
LSM 510 Meta (Carl Zeiss, Hertfordshire, U.K.). To further analyze the function of TXNIP in vitro, we
Coimmunoprecipitation. The interactions between SCAP and
SREBP-2, TXNIP, or Insig-1 proteins in HK-2 cells were analyzed by
transfected HK-2 cells with control vectors or TXNIP siRNA
coimmunoprecipitation (Co-IP). In brief, the cell IP lysis buffer plasmids, followed by 48 h of AGE-BSA treatment. TXNIP
(Thermo Scientific Pierce) was adopted for extracting total protein. deletion within HK-2 cells was confirmed through Western
Cell lysates were cleared by centrifugation, and supernatants were blot analysis (Figure 1C). Then, we conducted Oil Red O
immunoprecipitated with the appropriate antibodies using protein A/ staining. The lipid droplet accumulation degree in the AGE-
G-agarose beads. Next, the samples were subjected to immunoblot- BSA-treated HK-2 cells increased relative to control (Figure
ting analysis with the indicated antibodies. 2C). This result was confirmed using the results of the
Statistical Analysis. Results were analyzed with SPSS20.0 and cholesterol quantitative assay (Figure 2D). However, the
expressed in the form of mean ± S.E.M. One-way ANOVA and then silencing of TXNIP reduced the intracellular cholesterol and
Bonferroni’s multiple comparison tests were adopted for multiple
comparisons. P values <0.05 indicated statistical significance. lipid droplet contents within HK-2 cells exposed to AGE-BSA


treatment (Figure 2C,D).
Silencing of TXNIP Reduced HMGCoAR, LDLr,
RESULTS nSREBP-2, and SCAP Expression In Vivo and In Vitro.
Biochemical Features of the Mice. The basic mouse To explore the mechanism by which TXNIP regulated the
features when the experiment is completed are presented in tubular cholesterol metabolism at the molecular level, we
Table 1. As a result, the diabetic mice showed higher FBG and analyzed LDLr, HMGCoAR, SCAP, SREBP-2, and nSREBP-2
the db/db mice had increased body weight, AGEs, TG, and levels within HK-2 cells and the db/db mouse kidneys.
TC levels compared with the db/m mice. Relative to those in According to Figure 3A, the db/db mice had increased LDLr,
the db/db mice, the levels of FBG and AGEs in the db/db HMGCoAR, SCAP, SREBP-2, and nSREBP-2 protein levels
mice exposed to high-AGE diets were significantly elevated. within the kidneys relative to those in the db/m mice. The
However, silencing of TXNIP exhibited a remarkable decrease renal levels of these proteins in the db/db mice exposed to
C https://doi.org/10.1021/acs.jafc.1c03559
J. Agric. Food Chem. XXXX, XXX, XXX−XXX
Journal of Agricultural and Food Chemistry pubs.acs.org/JAFC Article

indicating that the reduced expression of nSREBP-2 might be


due to the attenuated ER-to-Golgi apparatus transport of the
SCAP−SREBP-2 complexes.
Silencing of the TXNIP Reduced the Translocation of
SCAP−SREBP-2 Complexes In Vitro. To explore the above
speculation, this study conducted confocal microscopy to
analyze the translocation of SCAP from ER-to-Golgi apparatus
within HK-2 cells. As a result, loading with AGE-BSA
promoted the deposition of SCAP within the Golgi apparatus,
but this effect was notably inhibited by the silencing of TXNIP
(Figure 4A). However, the results of Co-IP showed a lack of
interaction between TXNIP and SCAP−SREBP-2 protein
complexes, although the number of SCAP−SREBP-2 protein
complexes was significantly higher in the AGE-BSA-treated
HK-2 cells (Figure 4B). Based on the above findings, TXNIP
does not affect the transport of SCAP−SREBP-2 protein
complexes by directly binding to the complexes. The enhanced
transport of the complexes might be related to the increased
number of bonds between SREBP-2 and SCAP after
upregulating SCAP and SREBP-2. However, silencing of
TXNIP weakened the transport of SCAP (Figure 4A) and
reduced the interaction between SCAP and SREBP-2 (Figure
4C), indicating that TXNIP silencing attenuated the ER-to-
Golgi apparatus transport of SCAP−SREBP-2 protein
complexes, which reduced the activation of nSREBP-2,
followed by the downregulation of HMGCoAR and LDLr
proteins.
Silencing of TXNIP Increased Insig-1 Expression In
Vivo and In Vitro. Given that Insig-1 has an important role in
the transport of SCAP−SREBP-2 complexes, we further
evaluated the expression of Insig-1 within HK-2 cells and
kidneys of the db/db mice. It was found that the protein level
of Insig-1 decreased within the kidneys of the db/db mice
relative to those in the db/m mice. The protein levels of Insig-
1 in the kidneys of the high-AGE-diet-fed db/db mice
markedly declined compared with those fed with basal diet.
Nonetheless, the silencing of TXNIP clearly upregulated Insig-
Figure 1. AGEs increased TXNIP expression in the kidneys of 1 protein of both groups (Figure 5A). Furthermore, we
diabetic mice and HK-2 cells. The expression of TXNIP protein in the observed the low Insig-1 protein level within the AGE-BSA-
kidneys of mice (A, B) and in HK-2 cells (C) was determined by IHC treated HK-2 cells and the silencing of TXNIP upregulated
staining and Western blotting. Image J was used to quantify the Insig-1 (Figure 5B). Additionally, Insig-1 physically interacted
relative levels of proteins. GAPDH was used as an internal control. with SCAP−SREBP-2 protein complexes in the HK-2 cells,
Values are expressed as the mean ± S.E.M. of five independent and AGE-BSA inhibited the interaction between Insig-1 and
experiments. **P < 0.01 versus the db/m group or the control cells
(Ctr). #P < 0.05 versus db/db group, and ##P < 0.01 versus AGE-BSA- SCAP−SREBP-2 protein complexes (Figure 5C). The
treated cells.▲▲P < 0.01 versus AGEs + db/db group. inhibitory effect may be ascribed to the low expression of
Insig-1. However, silencing of TXNIP did not obviously
change the interaction between SCAP and Insig-1 (Figure 5D),
high-AGE diet markedly increased relative to those exposed to suggesting that silencing of TXNIP reduced the ER-to-Golgi
the basal diet. Nonetheless, silencing of TXNIP clearly reduced apparatus transport of the SCAP−SREBP-2 complexes but did
the expression levels of HMGCoAR, LDLr, nSREBP-2, and not increase their retention in the ER in spite of the
SCAP but did not reduce SREBP-2 expression in the db/db upregulation of Insig-1, which might be due to the down-
mice exposed to high-AGE and basal diets, respectively. In the regulation of SCAP.
in vitro study, the HMGCoAR, LDLr, SREBP-2, nSREBP-2, Silencing of TXNIP Improved the Renal Morphology
and SCAP protein expressions within AGE-BSA-treated HK-2 and Function in the Diabetic Mice. We also examined the
cells considerably increased relative to control. These proteins role of TXNIP on the structure and function of the kidneys of
can be suppressed by silencing TXNIP, other than SREBP-2 diabetic mice. First, the results of HE staining indicated no
(Figure 3B). These results suggested that the upregulation of obvious pathological change in the glomeruli and tubules in
SCAP and SREBP-2 induced by AGEs may promote SCAP− the db/m mice. Vacuolar degeneration of renal tubular
SREBP-2 complexes to transport to Golgi apparatus, where epithelial cells was observed within the db/db mouse kidneys.
SREBP-2 was hydrolyzed. Its active fragment (nSREBP-2) Such an alteration was pronounced in the db/db mice exposed
entered the nucleus and increased the expression levels of to high-AGE diet relative to those fed with basal diet.
HMGCoAR and LDLr. However, TXNIP silencing decreased However, the silencing of TXNIP improved the morphology of
nSREBP-2 expression without affecting the SREBP-2 level, the renal tubules (Figure 6A). The results of PAS staining
D https://doi.org/10.1021/acs.jafc.1c03559
J. Agric. Food Chem. XXXX, XXX, XXX−XXX
Journal of Agricultural and Food Chemistry pubs.acs.org/JAFC Article

Figure 2. Silencing of TXNIP reduced renal tubular lipid accumulation in vivo and in vitro. Renal Oil Red O staining and the semiquantitative
analysis for the percent of the positive areas in each group (A). The intracellular cholesterol contents in the kidneys of mice (B). Oil Red O staining
and the positive percentage of HK-2 cells (C). The intracellular cholesterol content in HK-2 cells (D). Oil Red O staining was observed under a
light microscope (400×). The percentage of positive staining areas in the kidneys of mice was semiquantitative analyzed by Image J. The positive
percentage of HK-2 cells was counted from five experiments. Values of intracellular cholesterol content are expressed as the mean ± S.E.M. of five
independent experiments. **P < 0.01 versus the db/m group or the control cells (Ctr). #P < 0.05 and ##P < 0.01 versus db/db group or AGE-BSA-
treated cells. ▲P < 0.05 and ▲▲P < 0.01 versus the AGEs + db/db group.

showed that the glomerular vascular loop was clear in the db/ among the db/db mice were elevated. Additionally, for the db/
m mice, along with a normal basement membrane. Mesangial db mice fed on a high-AGEs diet, the above parameters
expansion in the renal glomeruli was detected among the db/ showed significantly elevated levels relative to those in the db/
db mice, and this change was more significant within the db/ db mice, while no obvious change in BUN was observed in
db mouse kidneys fed with high-AGE diet compared with both groups. However, TXNIP silencing remarkably decreased
those fed with basal diet. However, TXNIP silencing these parameters compared with those in the db/db mice or
significantly alleviated these pathological changes (Figure
those fed with high-AGE diet. These results suggested that
6B). As revealed by Masson staining, collagen fibers
AGEs caused the morphological and functional injuries of renal
significantly accumulated within tubular interstitium in the
db/db mice (stained blue), especially in the tubular glomeruli and tubules in diabetic mice. However, TXNIP
interstitium of high-AGE diet-fed db/db mice, while TXNIP silencing greatly improved these changes. This effect might be
silencing significantly reduced the deposition of collagen fibers related to the decreased level of lipid deposition in the kidney,
(Figure 6C). Furthermore, compared with the db/m mice, although other mechanisms may be involved given that TXNIP
SCR (Figure 6D), BUN (Figure 6E), u-NGAL (Figure 6F), deletion can improve renal inflammatory response and
β2-MG (Figure 6G), and 24 h urine protein (Figure 6H) levels oxidative stress.25,26
E https://doi.org/10.1021/acs.jafc.1c03559
J. Agric. Food Chem. XXXX, XXX, XXX−XXX
Journal of Agricultural and Food Chemistry pubs.acs.org/JAFC Article

Figure 3. Silencing of TXNIP reduced HMGCoAR, LDLr, nSREBP-2, and SCAP expression in the kidneys of diabetic mice and HK-2 cells. The
protein expression of HMGCoAR, LDLr, SREBP-2, nSREBP-2, and SCAP in the kidneys of mice (A) and in HK-2 cells (B) was determined by
Western blotting. Image J was used to quantify the relative levels of proteins. GAPDH or Lamin A was used as an internal control. Values are
expressed as the mean ± S.E.M. of five independent experiments. *P < 0.05 and **P < 0.01 versus the db/m group or the control cells (Ctr). #P <
0.05 and ##P < 0.01, versus db/db group or AGE-BSA-treated cells. ▲P < 0.05 and ▲▲P < 0.01 versus the AGEs + db/db group. ns = not
significant, versus db/db group, AGEs + db/db group or AGE-BSA-treated cells.

F https://doi.org/10.1021/acs.jafc.1c03559
J. Agric. Food Chem. XXXX, XXX, XXX−XXX
Journal of Agricultural and Food Chemistry pubs.acs.org/JAFC Article

Figure 4. Interactions among SCAP, SREBP-2, and TXNIP proteins.


The translocation of GFP-SCAP from the ER to the Golgi was
investigated using confocal microscopy after staining with an anti- Figure 5. Expression of Insig-1 proteins and the interactions among
Golgi antibody. Silencing of TXNIP inhibited the translocation of Insig-1, SREBP-2, and SCAP proteins. The protein expression of
GFP-SCAP from the ER to the Golgi in HK-2 cells (A). The Insig-1 in the kidneys of mice (A) and in HK-2 cells (B) was
interaction between SCAP and SREBP-2 and TXNIP proteins was determined by Western blotting. Image J was used to quantify the
investigated using Co-IP. AGE-BSA increased the interactions of relative levels of proteins. GAPDH was used as an internal control.
SCAP and SREBP-2 in HK-2 cells, whereas TXNIP protein was not Values are expressed as the mean ± S.E.M. of five independent
detected (B). Silencing of TXNIP reduced the interaction between experiments. **P < 0.01 versus the db/m group or the control cells
SCAP and SREBP-2 (C). (Ctr). #P < 0.05 and ##P < 0.01 versus db/db group or AGE-BSA-
treated cells. ▲P < 0.05 versus the AGEs + db/db group. Interactions


among Insig-1, SREBP-2, and SCAP proteins were investigated using
DISCUSSION Co-IP. AGE-BSA increased the interactions of SCAP−SREBP-2
protein complexes in HK-2 cells and reduced the interactions between
The fatty kidney is an important risk factor for renal injury.27 SCAP and Insig-1 (C). Silencing of TXNIP did not change the
Therefore, reducing renal lipid accumulation is considered an interaction between SCAP and Insig-1 (D).
effective means of preventing the development of DKD. In a
previous study, we showed that AGEs can cause abnormal
cholesterol metabolism in the renal tubules of T2DM rats. The Then, SREBP-2 hydrolysis was inhibited in the Golgi
current study first examined the role of TXNIP in the apparatus, followed by reduced cholesterol production
accumulation of lipids in the kidneys of high-AGE diet-fed regulated by HMGCoAR and cholesterol absorption regulated
diabetic mice. A functional study was conducted on AGE- by LDLr.
treated HK-2 cells. As a result, TXNIP accounts for an As suggested by more and more studies, TXNIP has a key
important regulator for lipid deposition within renal tubular function in lipid metabolism.28 Recently, Chunyang Du et al.
cells under high-AGE conditions. TXNIP silencing remarkably reported that TXNIP deficiency alleviates renal fatty acid
inhibited AGE-induced cholesterol accumulation by down- metabolism in mice with streptozotocin-induced type 1
regulating the expression of SCAP, thus inhibiting the ER-to- diabetes.29 However, to date, only few studies have analyzed
Golgi apparatus transport of SCAP−SREBP-2 complexes. the TXNIP’s function in the metabolism of cholesterol in the
G https://doi.org/10.1021/acs.jafc.1c03559
J. Agric. Food Chem. XXXX, XXX, XXX−XXX
Journal of Agricultural and Food Chemistry pubs.acs.org/JAFC Article

Figure 6. continued

expressed as the mean ± S.E.M. of five mice in each group. Positive


fibrotic tissue stained by Masson staining was quantified by image
analysis using Image J software, presented as the percentage of the
total area. The histogram is expressed as the mean ± S.E.M. of five
mice in each group. The levels of SCR (D), BUN (E), u-NGAL (F),
β2-MG (G), and 24 h urine protein (H) in mice. Values are expressed
as the mean ± S.E.M. of five independent experiments. *P < 0.05 and
**P < 0.01 versus the db/m group. #P < 0.05 and ##P < 0.01 versus
db/db group. ▲P < 0.05 and ▲▲P < 0.01 versus the AGEs + db/db
group. ns = not significant, versus db/db group.

kidney. The present work suggested the role of TXNIP as a key


regulatory factor for the metabolism of cholesterol within the
kidneys of T2DM mice as well as within the cultured HK-2
cells exposed to high-AGE conditions. Renal TXNIP level was
dramatically upregulated in the diabetic mice relative to that in
the control mice, and the amounts of lipid droplets and
intracellular cholesterol levels in the kidneys increased.
Moreover, renal pathological changes and decreased renal
function were observed, especially in the renal tubules. These
effects were more pronounced in the high-AGE-diet-fed
diabetic mice. TXNIP silencing remarkably decreased these
variables. Meanwhile, the TXNIP protein level was upregulated
by AGEs in the HK-2 cells, and lipid droplet accumulation
resulted from AGEs. However, TXNIP silencing remarkably
suppressed the above process within the HK-2 cells. Based on
the above findings, TXNIP was related to cholesterol
deposition within the renal tubules of diabetic mice.
To further investigate the mechanism governing the effect of
TXNIP on the fatty kidney, we investigated the effect of
TXNIP on the feedback regulation of cholesterol in the HK-2
cells and kidneys of T2DM mice. We observed that the renal
protein levels of HMGCoAR, LDLr, SCAP, nSREBP-2, and
SREBP-2 increased in diabetic mice, especially in the kidneys
of the high-AGE-diet-fed diabetic mice. Similar results were
found in the AGE-treated HK-2 cells. However, TXNIP
silencing clearly reduced HMGCoAR, LDLr, nSREBP-2, and
SCAP protein expression, in addition to SREBP-2, in the
kidneys of diabetic mice and high-AGE-diet-fed diabetic mice,
as well as AGE-treated HK-2 cells. Further studies showed that
loading with AGE-BSA accelerated SCAP−SREBP-2 complex
formation while promoting their transportation to the Golgi
apparatus. However, these could be inhibited by TXNIP
silencing in the HK-2 cells. The above results suggested that
AGEs facilitated the ER-to-Golgi apparatus transport of
SCAP−SREBP-2 complexes by upregulating SREBP-2 and
SCAP expressions. Thus, SREBP-2 hydrolysis was promoted.
Then, nSREBP-2 fragments that entered the nucleus increased
in number and were bound to the promoters of HMGCoAR
and LDLr, thus enhancing the transcription and translation of
HMGCoAR and LDLr, leading to increased cholesterol
synthesis and uptake. However, TXNIP silencing decreased
the expression of SCAP and attenuated ER-to-Golgi apparatus
translocation of SCAP−SREBP-2 complexes, thus down-
regulating the expression of HMGCoAR and LDLr and
Figure 6. Silencing of TXNIP improved renal morphology and
ultimately reducing renal tubular cholesterol synthesis and
function of the diabetic mice. HE (A), PAS (B), and Masson (C)
staining in the kidneys of mice was observed under a light microscope uptake. Until now, there is no report on the relationship
(200×). The mesangial area was expressed quantitatively by between TXNIP and SCAP. Previous studies have shown that
calculating the percentage of the total glomerular area that was PAS glycosylation of SCAP prolongs its half-life and enhances the
positive. Fifteen glomerular tufts per animal were chosen randomly for stability of SCAP protein.30 Therefore, we suspect that
analysis. The histogram represents the mesangial matrix index, silencing TXNIP may affect the glycosylation of SCAP,
H https://doi.org/10.1021/acs.jafc.1c03559
J. Agric. Food Chem. XXXX, XXX, XXX−XXX
Journal of Agricultural and Food Chemistry pubs.acs.org/JAFC Article

resulting in the accelerated degradation of SCAP. However, Authors


further research is warranted to verify the above speculation. Juan Chen − Department of Endocrinology, Jiangsu Province
Insig-1 is a membrane protein that resides within ER, which Hospital of Chinese Medicine, Affiliated Hospital of Nanjing
has an important function in regulating SREBP proteolytic University of Chinese Medicine, Nanjing 210000, China
processing.31 It is ubiquitinated and degraded rapidly by Lili Sun − Department of Endocrinology and Metabolism, The
proteasomes when cholesterol is depleted in the cells under First Affiliated Hospital of Soochow University, Suzhou,
physiological conditions. Conversely, the ubiquitination of Jiangsu 215000, China
Insig-1 is blocked, and the protein is stabilized when Bimin Shi − Department of Endocrinology and Metabolism,
intracellular cholesterol is accumulated in cells.32 However, The First Affiliated Hospital of Soochow University, Suzhou,
cells come round the SREBP cholesterol suppression Jiangsu 215000, China
mechanism by modulating Insig-1 protein expression upon
Complete contact information is available at:
ER stress or hypotonic shock. In the current study, we found
https://pubs.acs.org/10.1021/acs.jafc.1c03559
that the renal protein level of Insig-1 decreased in the diabetic
mice, especially in high-AGE-diet-fed diabetic mice. Similar
Author Contributions
results were found in the AGE-treated HK-2 cells. ⊥
Furthermore, the results of Co-IP showed that AGE-BSA H.S. and J.C. contributed equally to this work. H.S. designed
inhibited the interaction between Insig-1 and SCAP−SREBP-2 the experiments. J.L., B.S., J.C., and L.S. performed the
protein complexes. These results implied that AGEs facilitated experiments. H.S., J.C., and J.L. revised the manuscript. All
the ER-to-Golgi apparatus transport of SCAP−SREBP-2 authors read and approved the final manuscript.
complexes by downregulating the expression of Insig-1 but Notes
upregulating that of SREBP-2 and SCAP, which enhanced ER- The authors declare no competing financial interest.


to-Golgi apparatus delivery of the SCAP−SREBP-2 complexes.
Hence, the transcriptional rates of SREBP-2 target genes
ACKNOWLEDGMENTS
increased, thereby leading to an increase in cholesterol
synthesis and uptake. However, TXNIP silencing did not This work was supported by grants from the National Natural
increase SCAP−SREBP-2 complex retention within ER in HK- Science Foundation of China (grant no. 81700632 to H.S.),
2 cells, despite the clearly elevated expression of the Insig-1 the Natural Science Foundation of Jiangsu Province (grant no.
protein, suggesting that the effect of TXNIP on the BK20170366 to H.S.), People’s Livelihood Science and
transportation of complexes was mainly due to its regulation Technology of Suzhou (grant no. SYS2020104 to H.S., grant
of SCAP rather than Insig-1. In addition, previous studies have no. SS202008 to B.S.), and China Postdoctoral Science
shown that binding of HMGCoAR to Insig-1 proteins leads to Foundation (2020M671559 to J.C.).


the ubiquitination and degradation of HMGCoAR,33 indicat-
ing that the upregulation of Insig-1 induced by TXNIP REFERENCES
silencing may further promote the combination of Insig-1 and
HMGCoAR, which promotes the degradation of HMGCoAR (1) Xu, Y.; Wang, L.; He, J.; Bi, Y.; Li, M.; Wang, T.; Jiang, Y.; Dai,
M.; Lu, J.; Xu, M.; Li, Y.; Hu, N.; Li, J.; Mi, S.; Chen, C. S.; Li, G.;
and inhibits cholesterol synthesis.
Mu, Y.; Zhao, J.; Kong, L.; Chen, J.; Lai, S.; Wang, W.; Zhao, W.;
To sum up, this study suggests the key function of TXNIP in Ning, G.; et al. Prevalence and control of diabetes in Chinese adults.
regulating lipid accumulation in the kidneys of diabetic mice JAMA, J. Am. Med. Assoc. 2013, 310, 948−959.
fed with high-AGE diet as well as in HK-2 cells exposed to (2) de Vries, A. P.; Ruggenenti, P.; Ruan, X. Z.; Praga, M.; Cruzado,
AGE treatment. The AGEs can promote SREBP-2 and SCAP J. M.; Bajema, I. M.; D’Agati, V. D.; Lamb, H. J.; Pongrac Barlovic, D.;
levels while downregulating Insig-1 expression, thus enhancing Hojs, R.; Abbate, M.; Rodriquez, R.; Mogensen, C. E.; Porrini, E.
the ER-to-Golgi apparatus translocation of SCAP−SREBP-2 Fatty kidney: emerging role of ectopic lipid in obesity-related renal
complexes, where SREBP-2 is hydrolyzed. The active frag- disease. Lancet Diabetes Endocrinol. 2014, 2, 417−426.
ments of SREBP-2 (nSREBP-2) enter the nucleus, thereby (3) Yokoo, T.; Clark, H. R.; Pedrosa, I.; Yuan, Q.; Dimitrov, I.;
upregulating HMGCoAR and LDLr expression levels and Zhang, Y.; Lingvay, I.; Beg, M. S.; Bobulescu, I. A. Quantification of
ultimately increasing the cholesterol synthesis and uptake. renal steatosis in type II diabetes mellitus using dixon-based MRI. J.
However, TXNIP silencing alleviates diabetic renal tubular Magn. Reson. Imaging 2016, 44, 1312−1319.
(4) Wang, Y. C.; Feng, Y.; Lu, C. Q.; Ju, S. Renal fat fraction and
lipid accumulation by inhibiting cholesterol absorption
diffusion tensor imaging in patients with early-stage diabetic
induced by LDLr and cholesterol production regulated by nephropathy. Eur. Radiol. 2018, 28, 3326−3334.
HMGCoAR through the downregulation of SCAP expression (5) Herman-Edelstein, M.; Scherzer, P.; Tobar, A.; Levi, M.; Gafter,
and subsequent inhibited ER-to-Golgi apparatus translocation U. Altered renal lipid metabolism and renal lipid accumulation in
of SCAP−SREBP-2 complexes. Therefore, TXNIP is a human diabetic nephropathy. J. Lipid Res. 2014, 55, 561−572.
potential therapeutic target for the diabetic fatty kidney. (6) Fufaa, G. D.; Weil, E. J.; Nelson, R. G.; Hanson, R. L.;

■ AUTHOR INFORMATION
Corresponding Authors
Bonventre, J. V.; Sabbisetti, V.; Waikar, S. S.; Mifflin, T. E.; Zhang, X.;
Xie, D.; Hsu, C. Y.; Feldman, H. I.; Coresh, J.; Vasan, R. S.; Kimmel,
P. L.; Liu, K. D. Association of urinary KIM-1, L-FABP, NAG and
NGAL with incident end-stage renal disease and mortality in
Hong Sun − Department of Endocrinology and Metabolism, American Indians with type 2 diabetes mellitus. Diabetologia 2015,
The First Affiliated Hospital of Soochow University, Suzhou, 58, 188−198.
Jiangsu 215000, China; orcid.org/0000-0003-4335- (7) Kim, S. S.; Song, S. H.; Kim, I. J.; Kim, W. J.; Jeon, Y. K.; Kim, B.
2789; Email: sunhong_611@126.com H.; Kwak, I. S.; Lee, E. K.; Kim, Y. K. Nonalbuminuric proteinuria as a
Jianzhong Li − Department of Nephrology, The First Affiliated biomarker for tubular damage in early development of nephropathy
Hospital of Soochow University, Suzhou, Jiangsu 215000, with type 2 diabetic patients. Diabetes/Metabolism Res. Rev. 2014, 30,
China; Email: ljzsnk@hotmail.com 736−741.

I https://doi.org/10.1021/acs.jafc.1c03559
J. Agric. Food Chem. XXXX, XXX, XXX−XXX
Journal of Agricultural and Food Chemistry pubs.acs.org/JAFC Article

(8) Yuan, Y.; Sun, H.; Sun, Z. Advanced glycation end products (25) Han, Y.; Xu, X.; Tang, C.; Gao, P.; Chen, X.; Xiong, X.; Yang,
(AGEs) increase renal lipid accumulation: a pathogenic factor of M.; Yang, S.; Zhu, X.; Yuan, S.; Liu, F.; Xiao, L.; Kanwar, Y. S.; Sun, L.
diabetic nephropathy (DN). Lipids Health and Dis. 2017, 16, No. 126. Reactive oxygen species promote tubular injury in diabetic
(9) Sun, H.; Yuan, Y.; Sun, Z. Update on Mechanisms of Renal nephropathy: The role of the mitochondrial ros-txnip-nlrp3 biological
Tubule Injury Caused by Advanced Glycation End Products. BioMed. axis. Redox Biol. 2018, 16, 32−46.
Res. Int. 2016, 2016, No. 5475120. (26) Ke, R.; Wang, Y.; Hong, S.; Xiao, L. Endoplasmic reticulum
(10) Zhang, Y.; Ma, K. L.; Ruan, X. Z.; Liu, B. C. Dysregulation of stress related factor IRE1alpha regulates TXNIP/NLRP3-mediated
the Low-Density Lipoprotein Receptor Pathway Is Involved in Lipid pyroptosis in diabetic nephropathy. Exp. Cell Res. 2020, 396,
Disorder-Mediated Organ Injury. Int. J. Biol. Sci. 2016, 12, 569−579. No. 112293.
(11) Engelking, L. J.; Cantoria, M. J.; Xu, Y.; Liang, G. (27) Ducasa, G. M.; Mitrofanova, A.; Fornoni, A. Crosstalk Between
Developmental and extrahepatic physiological functions of SREBP Lipids and Mitochondria in Diabetic Kidney Disease. Curr. Diabetes
pathway genes in mice. Semin. Cell Dev. Biol. 2018, 81, 98−109. Rep. 2019, 19, No. 144.
(12) Mahmood, D. F.; Abderrazak, A.; El Hadri, K.; Simmet, T.; (28) Alhawiti, N. M.; Al Mahri, S.; Aziz, M. A.; Malik, S. S.;
Rouis, M. The thioredoxin system as a therapeutic target in human Mohammad, S. TXNIP in Metabolic Regulation: Physiological Role
health and disease. Antioxid. Redox Signaling 2013, 19, 1266−1303. and Therapeutic Outlook. Curr. Drug Targets 2017, 18, 1095−1103.
(13) Thielen, L.; Shalev, A. Diabetes pathogenic mechanisms and (29) Du, C.; Wu, M.; Liu, H.; Ren, Y.; Du, Y.; Wu, H.; Wei, J.; Liu,
potential new therapies based upon a novel target called TXNIP. Curr. C.; Yao, F.; Wang, H.; Zhu, Y.; Duan, H.; Shi, Y. Thioredoxin-
Opin. Endocrinol., Diabetes Obes. 2018, 25, 75−80. interacting protein regulates lipid metabolism via Akt/mTOR
(14) Park, M. J.; Kim, D. I.; Lim, S. K.; Choi, J. H.; Kim, J. C.; Yoon, pathway in diabetic kidney disease. Int. J. Biochem. Cell Biol. 2016,
K. C.; Lee, J. B.; Lee, J. H.; Han, H. J.; Choi, I. P.; Kim, H. C.; Park, S. 79, 1−13.
H. Thioredoxin-interacting protein mediates hepatic lipogenesis and (30) Cheng, C.; Ru, P.; Geng, F.; Liu, J.; Yoo, J. Y.; Wu, X.; Cheng,
X.; Euthine, V.; Hu, P.; Guo, J. Y.; Lefai, E.; Kaur, B.; Nohturfft, A.;
inflammation via PRMT1 and PGC-1alpha regulation in vitro and in
Ma, J.; Chakravarti, A.; Guo, D. Glucose-Mediated N-glycosylation of
vivo. J. Hepatol. 2014, 61, 1151−1157.
SCAP Is Essential for SREBP-1 Activation and Tumor Growth.
(15) Andrews Guzman, M.; Arredondo Olguin, M.; Olivares
Cancer Cell 2015, 28, 569−581.
Gronhert, M. Glycemic control and oxidative stress markers and
(31) Dong, X. Y.; Tang, S. Q. Insulin-induced gene: a new regulator
their relationship with the thioredoxin interacting protein (TXNIP) in lipid metabolism. Peptides 2010, 31, 2145−2150.
gene in type 2 diabetic patients. Nutr. Hosp. 2015, 31, 1129−1133. (32) Lee, J. N.; Song, B.; DeBose-Boyd, R. A.; Ye, J. Sterol-regulated
(16) Yeh, W. J.; Yang, H. Y.; Pai, M. H.; Wu, C. H.; Chen, J. R. degradation of Insig-1 mediated by the membrane-bound ubiquitin
Long-term administration of advanced glycation end-product ligase gp78. J. Biol. Chem. 2006, 281, 39308−39315.
stimulates the activation of NLRP3 inflammasome and sparking the (33) Johnson, B. M.; De Bose-Boyd, R. A. Underlying mechanisms
development of renal injury. J. Nutr. Biochem. 2017, 39, 68−76. for sterol-induced ubiquitination and ER-associated degradation of
(17) Byon, C. H.; Han, T.; Wu, J.; Hui, S. T. Txnip ablation reduces HMG CoA reductase. Semin. Cell Dev. Biol. 2018, 81, 121−128.
vascular smooth muscle cell inflammation and ameliorates athero-
sclerosis in apolipoprotein E knockout mice. Atherosclerosis 2015, 241,
313−321.
(18) Mohamed, I. N.; Sarhan, N. R.; Eladl, M. A.; El-Remessy, A. B.;
El-Sherbiny, M. Deletion of Thioredoxin-interacting protein amelio-
rates high fat diet-induced non-alcoholic steatohepatitis through
modulation of Toll-like receptor 2-NLRP3-inflammasome axis:
Histological and immunohistochemical study. Acta Histochem. 2018,
120, 242−254.
(19) Wang, W.; Wang, C.; Ding, X. Q.; Pan, Y.; Gu, T. T.; Wang, M.
X.; Liu, Y. L.; Wang, F. M.; Wang, S. J.; Kong, L. D. Quercetin and
allopurinol reduce liver thioredoxin-interacting protein to alleviate
inflammation and lipid accumulation in diabetic rats. Br. J. Pharmacol.
2013, 169, 1352−1371.
(20) Wang, R.; Wang, Y.; Mu, N.; Lou, X.; Li, W.; Chen, Y.; Fan, D.;
Tan, H. Activation of NLRP3 inflammasomes contributes to
hyperhomocysteinemia-aggravated inflammation and atherosclerosis
in apoE-deficient mice. Lab. Invest. 2017, 97, 922−934.
(21) Meng, X. M.; Ren, G. L.; Gao, L.; Yang, Q.; Li, H. D.; Wu, W.
F.; Huang, C.; Zhang, L.; Lv, X. W.; Li, J. NADPH oxidase 4
promotes cisplatin-induced acute kidney injury via ROS-mediated
programmed cell death and inflammation. Lab. Invest. 2018, 98, 63−
78.
(22) Sowndhar Rajan, B.; Manivasagam, S.; Dhanusu, S.;
Chandrasekar, N.; Krishna, K.; Kalaiarasu, L. P.; Babu, A. A.;
Vellaichamy, E. Diet with high content of advanced glycation end
products induces systemic inflammation and weight gain in
experimental mice: Protective role of curcumin and gallic acid. Food
Chem. Toxicol. 2018, 114, 237−245.
(23) Sun, H.; Shao, X.; He, J.; Golos, M.; Shi, B. Role of the
mTORFOXO1 pathway in obesityassociated renal tubulointerstitial
inflammation. Mol. Med. Rep. 2019, 19, 1284−1293.
(24) Sun, H.; Sun, Z.; Varghese, Z.; Guo, Y.; Moorhead, J. F.;
Unwin, R. J.; Ruan, X. Z. Nonesterified free fatty acids enhance the
inflammatory response in renal tubules by inducing extracellular ATP
release. Am. J. Physiol.: Renal Physiol. 2020, 319, F292−F303.

J https://doi.org/10.1021/acs.jafc.1c03559
J. Agric. Food Chem. XXXX, XXX, XXX−XXX

You might also like