Anda di halaman 1dari 54

Author’s Accepted Manuscript

Vitamin C deficiency aggravates tumor necrosis factor


α-induced insulinresistance

Zhou Qing, Wu Xiao-Hui, Wu Xi-Mei, Zou Chao- Chun

www.elsevier.com/locate/ejphar

PII: S0014-2999(18)30210-3
DOI:
https://doi.org/10.1016/j.ejphar.2018.0
3.044 Reference: EJP71748
To appear in: European Journal of Pharmacology
Received date: 15 February 2018
Reviseddate: 26March2018
Accepted date: 29 March 2018
Cite this article as: Zhou Qing, Wu Xiao-Hui, Wu Xi-Mei
and Zou Chao-Chun, Vitamin C deficiency aggravates
tumor necrosis factor α-induced insulin r e s i s t a n c
e , European Journal of Pharmacology, https://doi.org/10.1016/j.ejphar.2018.03
This is a PDF file of an unedited manuscript that has
been accepted for publication. As a service to our
customers we are providing this early version of the
manuscript. The manuscript will undergo copyediting,
typesetting, and review of the resulting galley proof
before it is published in its final citable form. Please note
that during the production process errors may be
discovered which could affect the content, and all legal
disclaimers that apply to the journal pertain.
Vitamin C deficiency aggravates tumor necrosis
factor α-induced insulin resistance

Zhou Qinga: PhD, Wu Xiao-Huia: PhD, Wu Xi-Meib,


M.D., PhD, Zou Chao-Chuna, M.D.

aDepartment of Endocrinology of the affiliated


Children’s Hospital, b
Department of
Pharmacology, Zhejiang University School of
Medicine

Running title: Vitamin C in insulin resistance

Corresponding author:

Zou Chao-Chun, M.D., Professor

3333 Binsheng Road, Hangzhou 310051, China

Tel: +86-15067123060

E-mail: zcc14@zju.edu.cn Fax: +86-571-87033296


cardiovascular disease,
non-alcoholic fatty liver disease and cognitive
impairment in the metabolic syndrome (Bender et
al., 2013; Kim & Feldman, 2015; Wang et al.,
2017). The disturbance of energy nutrients
metabolism is a major characteristic of insulin
Abstract resistance. The liver plays a crucial role in the
maintenance of glucose homeostasis. Under normal
Chronic low-grade inflammation plays a major insulin action in the liver, the combination of
role in the development of insulin resistance. The insulin to its receptor on hepatocytes recruits and
potential role and underlying mechanism of vitamin activates insulin receptor substrate (IRS-1), which
C, an antioxidant and anti-inflammatory agent, was subsequently activates the downstream
investigated in tumor necrosis factor-α phosphatidylinositol 3-kinase (PI3K)/protein
(TNF-α)-induced insulin resistance. Gulonolactone kinase B (AKT) signaling pathway, and regulates
oxidase knockout (Gulo-/-) mice genetically unable gluconeogenesis and glycogenesis, in order to
to synthesize vitamin C were used to induce insulin maintain glucose homeostasis (Kim et al., 2015).
resistance by continuously pumping small doses of Chronic inflammation is a major contribution
TNF-α for seven days, and human liver to the development of insulin resistance
hepatocellular carcinoma cells (HepG2 cells) were (Nandipati et al., 2017; Xu et al., 2003). The two
used to induce insulin resistance by treatment with main inflammatory pathways, mitogen-activated
TNF-α. Vitamin C deficiency aggravated TNF-α- protein kinase (MAPKs) and nuclear factor-kappa
induced insulin resistance in Gulo-/- mice, B (NF-κB), play crucial roles in the progression of
resulting in worse glucose tolerance test (GTT) insulin resistance. MAPKs are a family of three
results, higher fasting plasma insulin level, and the distinct mammalian protein kinase termed c-Jun
inactivation of the protein kinase B NH2-terminal kinase (JNK), p38 and extracellular
(AKT)/glycogen synthase kinase-3β (GSK3β) signal-related kinase (ERK). The activation of
pathway in the liver. Vitamin C deficiency also MAPKs induces the serine phosphorylation of
worsened liver lipid accumulation and IRS-1, which result in the impairment of the
-/-
inflammation in TNF-α-treated Gulo mice. In downstream PI3K-AKT pathway, leading to a
HepG2 cells, vitamin C reversed the TNF-α- decrease in insulin signaling transduction
induced reduction of glucose uptake and glycogen (Fujishiro et al., 2003). The mediation of pro-
synthesis, which were mediated by increasing inflammatory cytokines in the activation of the
GLUT2 levels and the activation of the insulin inhibitor of IκB kinase (IKK)-β has been
receptor substrate (IRS-1)/AKT/GSK3β pathway. considered as a core mechanism that connects
Furthermore, vitamin C inhibited the TNF-α- metabolic inflammation and insulin resistance
induced activation of not only the mitogen- (Arkan et al., 2005). Studies have indicated that
activated protein kinase (MAPKs), but also nuclear TNF-α induces the activation of IKK-β, which
factor-kappa B directly targets the serine residue of IRS-1 or
(NF-κB) signaling. Taken together, vitamin C is rapidly removes tyrosine phosphate groups from
essential for preventing and improving insulin IRS-1 in a NF-κB-dependent manner, finally
resistance, and the supplementing with vitamin C leading to insulin resistance (Fernandez-Veledo et
may be an effective therapeutic intervention for al., 2006; Gao et al., 2002; Zabolotny et al., 2008).
metabolic disorders. Hence, inhibiting chronic inflammation may
Keywords: vitamin C; inflammatory cytokine; protect against insulin resistance.
insulin resistance; hepatocytes Vitamin C is a soluble antioxidant and
enzyme cofactor that participates in various
biological functions in vivo (Lykkesfeldt et al.,
2014). Vitamin C has been recognized as an anti-
inflammatory molecule due to its ability to
downregulate
pro-inflammatory cytokines induced by oxidative
1. Introduction stress (Burek & Rose, 2008; Gren, 2013). Studies
have shown that vitamin C supplementation
Insulin resistance is a principal feature of type- reduces the rate of congenital malformations in the
2 diabetes mellitus (Goldstein, 2002), which has offspring of diabetic rats (Siman & Eriksson,
been implicated in the development of 1997), and the daily intake of vitamin C attenuates
the exacerbation of cerebral ischemic injury in a
diabetic state (Iwata et al., 2014). Furthermore, All mice were housed at the Animal Care
low plasma vitamin C concentration has been Facility of Zhejiang University according to the
found to be associated with insulin resistance institutional guidelines for laboratory animals, and
(Donin et al., 2016). These evidences indicate that all the protocols were approved by the Zhejiang
vitamin C has a protective effect on diabetes, and University Institutional Animal Care and Use
vitamin C deficiency may be closely correlated to Committee.
the progression of insulin resistance. However, the 2.2 Tolerance test
direct effects of vitamin C on the amelioration of
insulin resistance remain unclear, and the At the end of the 7-day TNF-α exposure, an
underlying mechanism by which vitamin C intraperitoneal glucose tolerance test (GTT) was
protects against insulin resistance is far from clear. performed in mice. Mice were fasted overnight for
In the present study, we attempted to 14 h and intraperitoneally injected with glucose (1
determine whether vitamin C deficiency g/kg of body weight). Blood glucose
exacerbates TNF-α-induced insulin resistance in concentrations were measured from tail blood
Gulo-/- mice, and identify the underlying samples at 0, 30, 60 and 120 min after glucose
mechanism in human HepG2 cells, Focus was injection using a glucometer (Onetouch Ultra,
given on the link between TNF-α-associated Johnson).
inflammation and hepatic insulin resistance. 2.3 Blood and tissue sampling

On the next day of the GTT experiment, after


2. Materials and Methods fasting overnight (12 h), blood samples were
collected from the medial canthus of the eye using
2.1 Animals experiment a capillary tube, centrifuged at 1,400 g for 10 min,
and stored at -80 ºC. At the same time, liver tissues
Gulonolactone oxidase (Gulo-/-) knockout were surgically removed, portions of the liver
mice, which lack terminal enzyme Gulo in the tissue were immediately frozen in liquid nitrogen
synthesis pathway of vitamin C, is also absent in for subsequent biochemical analysis, and portions
humans, and depends completely on dietary of the liver tissue were fixed with 4%
vitamin C (Maeda et al., 2000), were obtained from formaldehyde in PBS for histopathological
the Mutant Mouse Regional Resource Center examination.
(University of California, Davis, CA). Serum 2.4 Biochemical analyses and histopathological
vitamin C levels were set within the normal or examinations
insufficient range by the supplementation of 3.3
g/L and 0.33 g/L of vitamin C in drinking water, Blood samples were used to carry out the
respectively. Then, 9-week-old male mice, which analysis of insulin using a mouse insulin enzyme-
were paired for body weight and glucose level, linked immunosorbent assays (ELISA) kit,
were randomly divided into four groups: LD group according to manufacturer’s instructions (R&D
(0.33 g/L of vitamin C), TNF-α + LD group (TNF- Systems, Minneapolis, USA). The homeostasis
α + 0.33 g/L of vitamin C), HD group (3.3 g/L of model assessment of insulin resistance (HOMA-
vitamin C), and TNF-α + HD group (TNF-α + 3.3 IR) index was calculated using the following
g/L of vitamin C); Each group comprised of eight formula: HOMA-IR= [FBS (mmol/l) × Fins
mice, and all mice were fed with a standard (mIU/l)] / 22.5. Hepatic glycogen levels were
laboratory diet in a measured with a glycogen assay kit (Biovision,
temperature-controlled (20-24°C) and humidity- USA), according to manufacturer’s instructions.
controlled (45-55%) environment. A 12 h/12 h Hepatic triglyceride and cholesterol contents were
light/dark cycle was maintained. At nine weeks, measured with a commercial assay kit (Nanjing
all mice had an ALZET osmotic pump (Durect, Jiancheng Bioengineering Institute, China),
Cupertino, CA, USA), which was implanted into according to manufacturer’s instructions. Hepatic
the interscapular subcutaneous space. Mice in the IL-1α and IL-6 levels were determined using
TNF-α treatment group were implanted with a mouse ELISA kits, according to manufacturer’s
pump filled with 6.44 μg/ml of TNF-α diluted in instructions (IL-6: eBioscience, CA; IL-1α:
the carrier (0.9% NaCl and 0.1% BSA), with a 7- Abcam, USA). The hematoxylin and eosin (H&E)
day pumping capacity and an infusion rate of 1 staining and immunohistochemistry staining of
μl/h. Mice in the control group were implanted NF-κBp65 were performed, as previously
with a pump filled with the carrier only. described (Ji et al., 2017). The primary antibody
against NF-κBp65 (1:800, CST, USA) was used in previously described (Tang et al., 2016). The
the experiment. Histological analyses were relative amounts of mRNA levels of the target gene
performed on five independent mice in a blinded were normalized to β-actin (for mice) or GAPDH
manner. (for human), and the differences in mRNA levels
2.5 Cell cultures were calculated using the 2-△△Ct method. The
primer sequences for the target genes (IL-1α, IL-6,
HepG2 cells were purchased from ATCC ACC1, FAS, PEPCK and G6P) used for real-time
(Manassas, VA) and maintained in Dulbecco's PCR are listed in Table 1 (Supplementary Table 1).
Modified Eagle Medium supplemented with 10% 2.8 Western blot
fetal bovine serum (Gibco, USA) and 1%
penicillin-streptomycin at 37°C with 5% CO 2. In Nuclear fractions from the mice liver
order to check insulin signaling molecules, cells homogenate and cytosolic, and membranal and
were treated with 100 nM of insulin for 10 min nuclear fractions from HepG2 cells were extracted
before the protein was collected, as previously using a Nuclear, Membrane and Cytoplasmic
reported (Lin & Lin, 2008; Nakajima et al., 2000; Protein Extraction Kit (Beyotime, China),
Zang et al., 2004). Cellular vitality was measured according to manufacturer's instructions. Next, 80
by cell counting kit-8 (Beyotime, China), μg of protein was separated by 8-10%
according to manufacturer’s instructions. Human SDS-PAGE, transferred onto a nitrocellulose
umbilical vein endothelial cells (HUVEC) were membrane, probed with antibodies at 4 °C
primary cultured and preserved in our laboratory, overnight, and incubated in IRDye 680 and 800
as described in a previous publication (Yao et al., second antibodies (Lincoln, Nebraska) for 1 h at
2015). The cells from passages 2 to 5 were used in room temperature. The immunoreactive signals
the experiment. were visualized using the Odyssey Infrared
Imaging System (LI-COR, Lincoln, NE).
2.6 Glycogen measurement, VEGF protein Antibodies against P-IKKα/β, IKKβ, P-JNK, JNK,
secretion and glucose uptake P-ERK, ERK, P-p38, p38, P-AKT, AKT,
P-GSK-3β, P-NF-κBp65, P-ACC and ACC were
Glycogen levels were measured in HepG2 all purchased from Cell Signal Technology
cells incubated with 100 nM of insulin for 3 h (USA). Antibodies against IκBα, P-IκBα, Sodium
using a glycogen assay kit (Biovision, USA), Potassium ATPase (Na+/K+-ATPase), GLUT2,
according to manufacturer’s instructions. IRS-1, P-IRS-1, VEGFA, PEPCK, G6P and FAS
In order to evaluate the VEGF protein were all
secretion, the HUVECs culture supernatant was obtained from Abcam (USA). Antibodies against
collected for determination of VEGF release using GAPDH and GSK-3β were purchased from Santa
a human VEGF Quantikine ELISA kit obtained Cruz Biotechnology (USA). Antibodies against α-
from R&D Systems (Minneapolis, USA), tubulin and Histone H3 were purchased from
according to manufacturer’s instructions. HuaAn Biotechnology (China). Histone H3,
For the glucose uptake measurement, the α-tubulin and Na+/K+-ATPase were used as the
medium of cells was replaced with DMEM (low- internal standards for the nuclear, cytosolic and
glucose, 1g/L) for 12 h, followed by DMEM (high- membranal protein, respectively. GAPDH was
glucose, 4.5g/L) containing 2-[N-(7-nitrobenz-2- used as the internal standard for the total protein.
oxa-1, 3-diazol-4-yl) amino]-2-deoxy-D-glucose
(2-NBDG, a fluorescent glucose analog, Sigma; 2.9 Immunofluorescent staining
100 μM final concentration) for 30 min. After
washing twice with PBS to remove free 2-NBDG, Cells were cultured in a 12-well plate covered
cells were collected. Then, 2-NBDG uptake with coverslips. After treatment, cells were fixed
activity was measured using a FACS flow with 4% paraformaldehyde for 10 min, followed by
cytometer (BD Biosciences, USA) and analyzed permeabilization with 0.1% Triton X-100 for 30
using the CellQuest software. min. After incubation with blocking buffer for
2.7 Quantitative real-time PCR 30min, the coverslips were incubated with primary
antibodies overnight at 4 °C. After washing with
Total RNA from HepG2 cells and mice liver TBST, cells were incubated with Alexa Fluor 546-
homogenates were extracted using TRIzol reagent tagged secondary antibody (Life Technologies).
(Takara Biotechnology, China), according to Then, the nuclei were counterstained with DAPI.
manufacturer’s instructions. The mRNA levels Imaging was performed using a Zeiss fluorescent
were determined by quantitative RT-PCR, as microscope equipped with an epifluorescence and
Axiocam camera system coupled with the [ACC1] and fatty acid synthase [FAS]), compared
axiovision software (Carl Zeiss, Oberkochen, with that in the TNF-α + HD group (Fig.2D).
Germany). These data demonstrate that vitamin C deficiency
2.10 Statistical analysis aggravated TNF-α-triggered lipid accumulation in
the liver of Gulo-/- mice.
Numerical data were expressed as mean ±
S.E.M. One-way ANOVA and Dunnett’s multiple 3.3 Vitamin C deficiency increased the TNF-α-
comparison tests were used to determine the induced inactivation of the
statistical significance. P<0.05 was considered
statistically significant. AKT/GSK3β pathway in Gulo-/- mice

In order to further explore the potential targets


3. Results of vitamin C on the amelioration of insulin
resistance, the specific protein expression of the
3.1 Vitamin C deficiency aggravated TNF-α- insulin signaling pathway in liver tissues of TNF-α-
induced insulin resistance in Gulo-/-mice treated Gulo-/- mice were analyzed. Figure 3 shows
that the phosphorylation of AKT and GSK3β
In order to determine the effect of vitamin C decreased in mice in the TNF-α + LD group, but
deficiency on glucose metabolism in TNF-α- significantly increased in mice in the TNF-α + HD
treated Gulo-/- mice, the fasting serum insulin level, group. These data suggest that vitamin C
hepatic glycogen level and hepatic mRNA deficiency aggravated insulin resistance by
expression of genes involved in gluconeogenesis increasing the TNF-α-induced inactivation of the
(phosphoenolpyruvate carboxykinase [PEPCK], AKT/GSK3β pathway in the liver of Gulo-/- mice.
glucose-6-phosphatase [G6P]) were assessed, and a 3.4 Vitamin C deficiency aggravated TNF-α-induced
GTT was performed. The plasma glucose levels of inflammation in Gulo-/- mice
Gulo-/- mice were higher in the TNF-α + LD group
(vitamin C deficiency group) than in the TNF-α + The production of pro-inflammatory cytokines
HD group (vitamin C normal group), especially at in the liver has been implicated in the development
30 and 60 min after glucose injection (Fig. 1A). of hepatic insulin resistance. In order to evaluate
The difference was indicated by the area under the the effect of vitamin C deficiency on TNF-α-
glucose curve (AUC) of the GTT (Fig. 1B). The triggered inflammation, the mRNA and protein
fasting serum insulin level and HOMA-IR of Gulo- level of IL-1α and IL-6 in liver tissues of Gulo-/-
/-
mice significantly increased in the TNF-α + LD mice were measured. The results revealed that the
group than in the TNF-α + HD group (Figs. 1C and mRNA and protein levels of IL-1α and IL-6
1D). Meanwhile, a decrease in glycogen level and significantly increased in the TNF-α + LD group
an increase in PEPCK and G6P mRNA levels were than that in the TNF-α + HD group (Figs. 4A and
found in liver tissues of Gulo-/- mice in the 4B). Moreover, immunohistochemistry revealed
TNF-α + LD group when compared with the TNF- that NF-κBp65 was widely expressed in the nuclei
α + HD group (Figs. 1E and 1F). These in vivo data of hepatocytes in mice in the TNF-α + LD group,
validates that vitamin C deficiency exacerbated when compared with the TNF-α + HD group (Fig.
insulin resistance in TNF-α-treated Gulo-/- mice. 4C), which was further confirmed by western blot
3.2 Vitamin C deficiency aggravated TNF-α- (Fig. 4D). These data indicate that TNF-α
triggered hepatic lipid accumulation in Gulo-/- treatment induced a significantly hepatic
mice inflammation, and vitamin C deficiency increased
Hepatic lipid metabolism is closely related the TNF-α-triggered inflammation in liver tissues
with hepatic insulin resistance. Hence, the hepatic of Gulo-/- mice.
lipid accumulation in these Gulo-/- mice was
examined. First, the liver histologic analysis from 3.5 Vitamin C improved TNF-α-induced
mice in the TNF-α + LD group indicated glucose and lipid metabolic disorder in HepG2
prominent vacuolization, when compared with cells
mice in the TNF-α + HD group (Fig. 2A). Since the animal models confirmed that
Consistent with the result from the H-&-E staining, vitamin C supplementation can improve insulin
livers from mice in the TNF-α + LD group resistance in TNF-α-treated Gulo-/- mice, the
exhibited increased triglyceride and cholesterol investigators subsequently explored the
content (Figs. 2B and 2C) and lipogenesis-related underlying mechanism by which vitamin C
gene mRNA expression (acetyl CoA carboxylase 1 supplementation improves insulin resistance.
Focus was given on the effects of vitamin C on expression were evaluated. The results revealed
glycogen synthesis, glucose uptake, and the that vitamin C significantly increased AKT
mRNA and protein levels of ACC1, FAS, PEPCK phosphorylation at position Ser473, GSK3β
and G6P. First, the cell viability of HepG2 treated phosphorylation at position Ser9, and IRS-1
with different concentrations of vitamin C for 24 or tyrosine phosphorylation at position Tyr632, and
48 h were quantified by cell counting kit-8 assays was accompanied by reduced IRS-1 serine
to exclude the cytotoxicity of vitamin C, and result phosphorylation at position Ser312, in
indicated that no cytotoxicity was observed after TNF-α-induced insulin resistant HepG2 cells (Fig.
exposure of HepG2 cells to vitamin C (100 μM, 6A). Moreover, vitamin C also reversed the TNF-
24h; Fig. 5A). Next, glycogen level, 2-NBDG α-mediated reduction of GLUT2 protein expression
uptake and mRNA and protein levels of ACC1, (Fig. 6B). The above results demonstrate that
FAS, PEPCK and G6P were tested, since these vitamin C can attenuate TNF-α-induced insulin
indicators are manifestations of insulin resistance. resistance by improving the IRS-1/AKT/GSK3β
The results revealed that vitamin C reversed the pathway and enhancing GLUT2 levels in HepG2
TNF-α-induced reduction of glycogen synthesis in cells.
a dose and time-dependent manner (Figs.5B and 3.7 Vitamin C upregulates the high glucose-
5C), and vitamin C also increased negated VEGF expression in HUVECs
TNF-α-impaired 2-NBDG uptake in HepG2 cells Diabetes mellitus is accompanied by a series
(Fig. 5D). Moreover, vitamin C inhibited the of diabetic macro-angiopathy and micro-
mRNA expression of ACC1, FAS, PEPCK and G6P angiopathy, such as atherosclerosis, retinopathy and
genes (Figs. 5E and 5F), downregulated the protein nephropathy. Furthermore, vascular endothelial
expression of PEPCK, G6P, FAS, and upregulated growth factor (VEGF) plays an important role in
the phosphorylation of ACC in HepG2 cells diabetic vascular complication. Therefore, the
induced by TNF-α (Fig. 5G), since effect of vitamin C on synthesis and the secretion
phosphorylation at 79 serine residue of ACC of VEGF in primary HUVEC incubated with high
inactivate the enzyme, indicating that vitamin C glucose for 24 h were explored. The results
inhibited gluconeogenesis and lipogenesis in revealed that high glucose inhibited VEGF protein
insulin resistant HepG2 cells treated by TNF-α. expression and secretion, while vitamin C
Taken together, these data demonstrate that upregulated the high glucose-negated VEGF
vitamin C improved glucose and lipid metabolic protein synthesis and secretion in HUVECs
disorders in TNF-α-induced insulin resistant (Supplementary Figure 1).
HepG2 cells. 3.8 Vitamin C inhibited the TNF-α-induced
3.6 Vitamin C restored TNF-α-negated activation of the IKKβ/IκBα/NF-κB pathway in
GLUT2 levels and the IRS-1/AKT/GSK3β HepG2 cells
pathway in HepG2 cells
IRS-1 phosphorylation plays a switch role for Insulin resistance has been considered as a
insulin signaling transduction. The serine chronic low-grade inflammatory response, and the
phosphorylation of IRS-1 is a negative regulator of activation of the IKKβ/NF-κB pathway is involved
the insulin-PI3K pathway. The serine in
phosphorylation of IRS-1 not only reduces TNF-α-induced insulin resistance. Hence, it was
tyrosine phosphorylation, but also accelerates IRS- further investigated whether vitamin C has an
1 degradation and the dissociation between insulin inhibitory effect on the activation of the IKKβ/NF-
receptor-IRS-1 and IRS-1-PI3K, leading to the κB signaling pathway.
inactivation of the AKT pathway (Moeschel et al., First, the time for the activation of the IKKβ/IκB-
2004; Yu et al., 2002). GSK3β phosphorylation in α/NF-κB pathway induced by TNF-α was
residue Ser9 plays a positive role in glycogen determined. The results revealed that TNF-α
synthesis, and GLUT2 is a predominant glucose induced IKKβ, IκB-α and NF-κB phosphorylation
transporter in the plasma membrane of maximally by 15 min, and declined by 60 min
hepatocytes. In order to explore whether vitamin C (Fig.7A).
reversed the TNF-α-mediated reduction of
glycogen synthesis involved in the activation of Furthermore, western blot analysis revealed that
the IRS-1/AKT/GSK3β pathway, and whether vitamin C blocked TNF-α-induced NF-κB
vitamin C improved the glucose uptake involved activation by inhibiting the p65 translocation from
in increasing GLUT2 level, the effects of vitamin cytoplasm to nuclei (Fig. 7B), and this inhibitory
C on the TNF-α-triggered inactivation of the IRS- effect of vitamin C on NF-κB translocation was
1/AKT/GSK3β pathway and GLUT2 protein also confirmed by immunofluorescence staining
(Fig.7C). Since vitamin C has an inhibitory effect of TNF-α for seven days, as previously described
on NF-κB translocation, the effect of vitamin C on (Chu et al., 2013; Zhao et al., 2015).
IKKβ and IκBα phosphorylation in the presence of
TNF-α was subsequently tested, which are at the The IKKβ/NF-κB pathway and MAP kinases
upstream of NF-κB. As shown in Figure 7D, JNK, p38 and ERK have been considered as the
vitamin C partially inhibited the two major transcription factors in response to
TNF-α-induced activation of IKKβ and IκBα. inflammatory cytokine stimuli. These kinases,
Taken together, these results clearly suggest that including IKKβ, JNK, p38 and ERK, have been
vitamin C can inhibit the IKKβ/IκBα/NF-κB activated by inflammatory cytokines, which
signaling pathway activated by TNF-α in HepG2 subsequently induce uncontrolled IRS serine
cells. phosphorylation, finally leading to insulin
3.9 Vitamin C inhibited the TNF-α-induced resistance (Tanti & Jager, 2009). Multiple studies
activation of JNK and p38 rather than ERK in have shown that JNK is a crucial mediator of
HepG2 cells insulin resistance: mice with JNK-deficient
Accumulating evidences have shown that the prevented insulin resistance induced by high fat
MAPK pathway is required for the TNF-α-induced diet (Han et al., 2013; Sabio et al., 2008); JNK
serine phosphorylation of IRS-1. Hence, the impact activity was significantly increased in liver,
of vitamin C on MAPK activation was investigated. muscle and adipose tissues in both dietary and
First, the time for the phosphorylation of MAPKs genetic (ob/ob) models of obesity; the absence of
activated by TNF-α was determined. The results JNK1 remarkably improved insulin sensitivity and
revealed that TNF-α increased JNK and p38 enhanced insulin receptor signaling capacity
phosphorylation maximally by 30 min, and (Hirosumi et al., 2002). In addition, the IKKβ/NF-
declined by 60 min (Fig.8A). κB pathway also play a major role in connecting
Furthermore, vitamin C significantly blocked the metabolic inflammation and insulin resistance: the
TNF-α-triggered phosphorylation of JNK and p38, deficiency of IKKβ in adipocytes markedly
but without inducing a significant inhibitory effect alleviated free fatty acid-induced inflammation
on ERK activation (Fig.8B). These results indicate and insulin resistance, while IKKβ overexpression
that vitamin C can inhibit the TNF-α-induced caused inflammation and insulin resistance (Jiao et
activation of JNK and p38 in HepG2 cells. al., 2011), and the overexpression of IκBα in the
liver or neurons of mice, which inhibited NF-κB
4. Discussion activation, attenuated high fat diet-induced body
weight gain and insulin resistance (Benzler et al.,
Obesity and diabetes mellitus have become a 2015; Wang et al., 2014). All these researches
major public problem worldwide, and both have demonstrate that JNK and the IKKβ/NF-κB
been considered as a chronic low-grade pathway are potential targets for the treatment of
inflammatory state. This insulin resistance.
low-grade inflammation is characterized by higher Vitamin C is an antioxidant with potent
levels of circulating cytokines and the activation of inhibitory action on multiple inflammatory
pro-inflammatory signaling pathways, which play signaling pathways. For example, Kyaw et al.
a central role in the pathogenesis of insulin (Kyaw et al., 2001) found that vitamin C inhibited
resistance (Khodabandehloo et al., 2016). JNK and p38 activation in rat aortic smooth
Inflammatory cytokines, such as TNF-α, IL-1 and muscle cells through its anti-oxidative property.
IL-6, disrupt insulin signaling at the insulin Several studies have also shown that vitamin C
receptor and IRS levels through multiple signaling inhibited the activation of the IκBα/NF-κB pathway
pathways, which finally contribute to the in primary HUVECs, protecting against
development of insulin resistance (Asrih & immunological hepatic injury in mice (Bowie &
Jornayvaz, 2013; Dali-Youcef et al., 2013). O'Neill, 2000; Carcamo et al., 2002; Liang et al.,
Cytokines can activate each other through the 2014). Similarly, the present data revealed that
inflammatory pathway, and TNF-α has been vitamin C inhibited the TNF-α-triggered activation
identified as one of the first links between low- of IKKβ, IκBα, NF-κB, JNK and p38 (although
grade inflammation and insulin resistance. Thus, without a significant inhibitory effect on ERK
in the present study, human HepG2 hepatocytes activation) in HepG2 cells, and inhibited the NF-
were treated with 10 ng/ml of TNF-α for 24 h to κB activation and the expression of IL-6 and IL-1α
induce insulin resistance, and the mouse model in liver tissues of
with insulin resistance was established by
continuously pumping small doses
gluconeogenesis, while ACC1 and FAS are
involved in the de novo lipogenesis. Decreased
PEPCK, G6P, ACC1 and FAS gene expression
were also observed in liver tissues obtained from
TNF-α-treated vitamin C normal Gulo-/- mice,
further supporting the improvement effect of
Gulo-/- mice. Therefore, vitamin C can be a good vitamin C on insulin resistance. Since GSK3β is an
strategy to target chronic inflammation linked to important factor in the regulation of hepatic
insulin resistance, and might play a protective role glycogenesis, the upregulation of the AKT/GSK3β
in the development of insulin resistance. A double- pathway in vitro and in vivo by vitamin C might
blind, randomized and placebo-controlled be correlated with its positive effect on glycogen
crossover trial revealed that vitamin C synthesis impaired by TNF-α.
supplementation decreased the risk for insulin GLUT2 is the major glucose transporter of
resistance under an environment of high exposure hepatocytes, which is responsible for glucose
to perfluorinated compounds, uptake during the postprandial phase and glucose
which were found to be positively associated with release during the fasting phase (Thorens, 2015).
insulin resistance (Kim et al., 2016). As a result, Multiple anti-diabetic drugs have been reported to
the present study investigated whether vitamin C recover the diminished level of hepatic GLUT2 in
deficiency exacerbate TNF-α-induced insulin mice fed on high fat diet (Souza-Mello et al.,
resistance in Gulo-/- mice. It was found that mice in 2010). Furthermore, a study revealed that the
the reduced GLUT2 level and glucose uptake in
TNF-α-treated vitamin C deficient group had higher insulin-resistant hepatocytes were recovered when
plasma glucose, increased fasting plasma insulin cells were co-treated with wax apple fruit extract
levels, decreased hepatic glycogen content, and (Shen et al., 2012). In agreement with the above
higher liver triglyceride and cholesterol levels, studies, GLUT2 levels and glucose uptake
while vitamin C supplementation significantly decreased in insulin-resistant HepG2 cells triggered
improved insulin sensitivity, abnormal glucose and by TNF-α, but this reductions have been reverted
lipid metabolism in when cells were co-treated with vitamin C. These
TNF-α-induced insulin-resistant Gulo-/- mice. results might partially explain the improvement
These results are in accordance with a previous effect of vitamin C on glucose tolerance in TNF-α-
study conducted by Aluwong, in which it was induced insulin-resistant Gulo-/- mice.
reported that vitamin C had a positive effect on VEGF is an angiogenic factor for endothelial
diabetes by ameliorating hyperglycemia, oxidative cells, which is strongly associated with the
stress and dyslipidemia in alloxan-induced development of diabetic vascular complications.
diabetic rats (Aluwong et al., 2016). Therefore, it VEGF level in response to high glucose stimuli is
was concluded that vitamin C deficiency differentially regulated in different types of cells or
exacerbates insulin resistance in tissues.
TNF-α-treated Gulo-/- mice, which might be The upregulation effect of high glucose stimulation
partially the result of losing the anti-inflammation on VEGF expression is observed in mesangial cells
property. and retinal pigment epithelial cells (Cai et al.,
At the cellular level, hepatic insulin resistance 2014; Kim et al., 2000), while this effect is the
is characterized by the impaired activation of the opposite observed in HUVECs (Yang et al., 2008),
IRS-1/PI3K/AKT signaling pathway. The animal indicating the differential role of VEGF in diabetes-
experiments have demonstrated that vitamin C associated vascular complications. Doronzo et al.
deficiency exacerbated TNF-α-induced hepatic reported that high glucose enhanced VEGF
insulin resistance and inflammation. This synthesis and secretion in primary vascular
prompted the investigators to determine whether smooth muscle cells (VSMC) from lean insulin-
vitamin C could inhibit inflammation-stimulated sensitive rats, but this effect was attenuated in
IRS-1 serine phosphorylation and restore the VSMC from obese insulin-resistant rats (Doronzo
impaired insulin signal transduction pathway. It et al., 2012), indicating that VEGF expression has
was found that vitamin C enhanced the activation a certain correlation with insulin resistance. In
of the IRS-1/AKT/GSK3β pathway, and order to explore the effect of vitamin C on VEGF
downregulated the expression of ACC1, FAS, expression, primary HUVECs treated with high
PEPCK and G6P in TNF-α-induced insulin glucose were cultured, and the results revealed
resistant HepG2 cells. This was because PEPCK that vitamin C upregulated the high glucose-
and G6P control the rate-limiting steps of liver inhibited VEGF protein synthesis and secretion in
HUVECs. This might be due to the anti-oxidative The authors declare no conflict of interest with
stress effect of vitamin C, since high glucose this work.
induces the generation of oxidative stress and
excessive oxidative stress could interfere with References
VEGF signaling (Yang et al., 2008). In vivo
studies have shown that vitamin C, through its Arkan, M.C., Hevener, A.L., Greten, F.R., Maeda, S., Li,
antioxidant effect, evidently increased capillary Z.W., Long, J.M., Wynshaw-Boris, A., Poli, G.,
density in ischemic gastrocnemius tissues of Olefsky, J. & Karin, M. (2005) IKK-beta links
diabetic rats, accompanied by the upregulation of inflammation to obesity-induced insulin resistance.
VEGF (El-Azab et al., 2012). These findings may Nat Med, 11, 191-198.
provide a certain basis for vitamin C in the Asrih, M. & Jornayvaz, F.R. (2013) Inflammation as a
treatment of diabetic vascular complication. potential link between nonalcoholic fatty liver
disease and insulin resistance. J Endocrinol, 218,
The limitation of the present study was that R25-36.
the ameliorative effect of vitamin C on Aluwong, T., Ayo, J.O., Kpukple, A. & Oladipo, O.O.
inflammation-associated insulin resistance in other (2016) Amelioration of Hyperglycaemia,
insulin responsive tissues such as adipose tissues Oxidative Stress and Dyslipidaemia in Alloxan-
and muscle was not examined. The effect of Induced Diabetic Wistar Rats Treated with
vitamin C on the vessel condition in insulin Probiotic and Vitamin C. Nutrients, 8.
resistant Gulo-/- mice and the positive effect of Bowie, A.G. & O'Neill, L.A. (2000) Vitamin C inhibits
vitamin C on other pro-inflammatory cytokine- NF-kappa B activation by TNF via the activation of
induced insulin resistance need to be clarified p38 mitogen-activated protein kinase. J Immunol,
through further studies. 165, 7180-7188.
Burek, C.L. & Rose, N.R. (2008) Autoimmune thyroiditis
and ROS. Autoimmun Rev, 7, 530-537.
5. Conclusions Bender, S.B., McGraw, A.P., Jaffe, I.Z. & Sowers, J.R.
(2013) Mineralocorticoid receptor-mediated
In summary, the present study demonstrates that vascular insulin resistance: an early contributor to
vitamin C inhibits diabetes-related vascular disease? Diabetes, 62,
313-319.
TNF-α-associated inflammation by targeting Benzler, J., Ganjam, G.K., Pretz, D., Oelkrug, R., Koch,
MAPKs and NF-κB signaling. Vitamin C C.E., Legler, K., Stohr, S., Culmsee, C., Williams,
supplementation prevents hepatic insulin resistance L.M. & Tups, A. (2015) Central inhibition of
caused by TNF-α. The effect of vitamin C in IKKbeta/NF-kappaB signaling attenuates high-fat
alleviating hepatic insulin resistance occurs in diet-induced obesity and glucose intolerance.
association with the prevention of TNF-α-induced Diabetes, 64, 2015-2027.
inflammation and corresponding impairment of Carcamo, J.M., Pedraza, A., Borquez-Ojeda, O. & Golde,
hepatic insulin signaling. Therefore, vitamin C D.W. (2002) Vitamin C suppresses TNF alpha-
might have potential for use in the treatment of induced NF kappa B activation by inhibiting I
metabolic disorders. kappa B alpha phosphorylation. Biochemistry, 41,
12995-13002.
Chu, J., Zhang, H., Huang, X., Lin, Y., Shen, T., Chen, B.,
Acknowledgments Man, Y., Wang, S. & Li, J. (2013) Apelin
ameliorates TNF-alpha-induced reduction of
This study was supported by the National glycogen synthesis in the hepatocytes through G
Natural Science Foundation of China (Grant protein-coupled receptor APJ. PLoS One, 8,
numbers 81170787 and 81670786), and the e57231.
Zhejiang Provincial Program for the Cultivation of Cai, Y., Li, X., Wang, Y.S., Shi, Y.Y., Ye, Z.,
High-Level Innovative Health Talents (2015). Yang, G.D., Dou, G.R., Hou, H.Y., Yang,
Conflict of interest
N., Cao, X.R. & Lu, Z.F. (2014) Hyperglycemia
promotes vasculogenesis in choroidal
neovascularization in diabetic mice by stimulating
VEGF and
SDF-1 expression in retinal pigment epithelial
cells. Exp Eye Res, 123, 87-96.
333-336.
Doronzo, G., Viretto, M., Russo, I., Mattiello, L., Anfossi, Han, M.S., Jung, D.Y., Morel, C., Lakhani, S.A.,
G. & Trovati, M. (2012) Effects of high glucose on Kim, J.K., Flavell, R.A. & Davis,
vascular endothelial growth factor synthesis and
secretion in aortic vascular smooth muscle cells R.J. (2013) JNK expression by macrophages
from obese and lean Zucker rats. Int J Mol Sci, 13, promotes obesity-induced insulin resistance and
9478-9488. inflammation. Science, 339, 218-222.
Dali-Youcef, N., Mecili, M., Ricci, R. & Andres, E. (2013) Iwata, N., Okazaki, M., Xuan, M., Kamiuchi, S.,
Metabolic inflammation: connecting obesity and Matsuzaki, H. & Hibino, Y. (2014) Orally
insulin resistance. Ann Med, 45, 242-253. administrated ascorbic acid suppresses neuronal
Donin, A.S., Dent, J.E., Nightingale, C.M., Sattar, damage and modifies expression of SVCT2 and
N., Owen, C.G., Rudnicka, A.R., GLUT1 in the brain of diabetic rats with cerebral
ischemia-reperfusion. Nutrients, 6, 1554-1577.
Perkin, M.R., Stephen, A.M., Jebb, S.A., Cook, Jiao, P., Ma, J., Feng, B., Zhang, H., Diehl, J.A., Chin,
D.G. & Whincup, P.H. (2016) Fruit, vegetable and Y.E., Yan, W. & Xu, H. (2011) FFA-induced
vitamin C intakes and plasma vitamin C: cross- adipocyte inflammation and insulin resistance:
sectional associations with insulin resistance and involvement of ER stress and IKKbeta pathways.
glycaemia in 9-10 year-old children. Diabet Med, Obesity (Silver Spring), 19, 483-491.
33, 307-315. Ji, X., Hu, X., Zou, C., Ruan, H., Fan, X., Tang, C., Shi,
El-Azab, M.F., Hazem, R.M. & Moustafa, Y.M. (2012) W., Mei, L., Zhu, H., Hussain, M., Zeng, L.,
Role of simvastatin and/or antioxidant vitamins in Zhang, X. & Wu, X. (2017) Vitamin C deficiency
therapeutic angiogenesis in experimental diabetic exacerbates diabetic glomerular injury through
hindlimb ischemia: effects on capillary density, activation of transforming growth
angiogenesis markers, and oxidative stress. Eur J factor-beta signaling. Biochim Biophys Acta,
Pharmacol, 690, 31-41. 1861, 2186-2195.
Fujishiro, M., Gotoh, Y., Katagiri, H., Sakoda, H.,
Ogihara, T., Anai, M., Onishi, Y., Ono, H., Abe, Kim, N.H., Jung, H.H., Cha, D.R. & Choi, D.S. (2000)
M., Shojima, N., Fukushima, Y., Kikuchi, M., Oka, Expression of vascular endothelial growth factor in
Y. & Asano, response to high glucose in rat mesangial cells. J
T. (2003) Three mitogen-activated protein kinases Endocrinol, 165, 617-624.
inhibit insulin signaling by different mechanisms in Kyaw, M., Yoshizumi, M., Tsuchiya, K., Kirima, K. &
3T3-L1 adipocytes. Mol Endocrinol, 17, 487-497. Tamaki, T. (2001) Antioxidants inhibit JNK and
Fernandez-Veledo, S., Nieto-Vazquez, I., Rondinone, p38 MAPK activation but not ERK 1/2 activation
C.M. & Lorenzo, M. (2006) Liver X receptor by angiotensin II in rat aortic smooth muscle cells.
agonists ameliorate TNFalpha-induced insulin Hypertens Res, 24, 251-261. Kim, O.K., Jun, W. &
resistance in murine brown adipocytes by Lee, J. (2015) Mechanism of ER Stress and
downregulating protein tyrosine phosphatase-1B Inflammation for
gene expression. Diabetologia, 49, 3038-3048. Hepatic Insulin Resistance in Obesity. Ann Nutr
Gao, Z., Hwang, D., Bataille, F., Lefevre, M., York, D., Metab, 67, 218-227.
Quon, M.J. & Ye, J. (2002) Serine phosphorylation
of insulin receptor substrate 1 by inhibitor kappa B Kim, B. & Feldman, E.L. (2015) Insulin resistance as a key
kinase complex. J Biol Chem, 277, 48115-48121. link for the increased risk of cognitive impairment
Goldstein, B.J. (2002) Insulin resistance as the in the metabolic syndrome. Exp Mol Med, 47,
core defect in type 2 diabetes mellitus. e149.
Khodabandehloo, H., Gorgani-Firuzjaee, S., Panahi, G. &
Am J Cardiol, 90, 3G-10G. Meshkani, R. (2016) Molecular and cellular
mechanisms linking inflammation to insulin
Gren, A. (2013) Effects of vitamin E, C and D resistance and beta-cell dysfunction. Transl Res,
supplementation on inflammation and oxidative 167, 228-256.
stress in streptozotocin-induced diabetic mice. Int J Kim, J.H., Park, H.Y., Jeon, J.D., Kho, Y., Kim,
Vitam Nutr Res, 83, 168-175 Hirosumi, J., S.K., Park, M.S. & Hong, Y.C. (2016)
Tuncman, G., Chang, L., Gorgun, C.Z., Uysal,
K.T., Maeda, K., Karin, The modifying effect of vitamin C on the
association between perfluorinated compounds and
M. & Hotamisligil, G.S. (2002) A central role for insulin resistance in the Korean elderly: a double-
JNK in obesity and insulin resistance. Nature, 420, blind, randomized, placebo-controlled crossover
trial. Eur J Nutr, 55, 1011-1020. Shen, S.C., Chang, W.C. & Chang, C.L. (2012) Fraction
Lin, C.L. & Lin, J.K. (2008) Epigallocatechin gallate from wax apple [Syzygium samarangense (Blume)
(EGCG) attenuates high glucose-induced insulin Merrill and Perry] fruit extract ameliorates insulin
signaling blockade in human hepG2 hepatoma cells. resistance via modulating insulin signaling and
Mol Nutr Food Res, 52, 930-939. inflammation pathway in tumor necrosis factor
Lykkesfeldt, J., Michels, A.J. & Frei, B. (2014) alpha-treated FL83B mouse hepatocytes. Int J Mol
Vitamin C. Adv Nutr, 5, 16-18. Sci, 13, 8562-8577.
Tanti, J.F. & Jager, J. (2009) Cellular mechanisms of
Liang, T., Chen, X., Su, M., Chen, H., Lu, G. & Liang, K. insulin resistance: role of stress-regulated serine
(2014) Vitamin C exerts beneficial kinases and insulin receptor substrates (IRS) serine
hepatoprotection against Concanavalin A-induced phosphorylation. Curr Opin Pharmacol, 9, 753-
immunological hepatic injury in mice through 762.
inhibition of NF-kappaB signal pathway. Food Thorens, B. (2015) GLUT2, glucose sensing and glucose
Funct, 5, 2175-2182. homeostasis. Diabetologia, 58, 221-232.
Maeda, N., Hagihara, H., Nakata, Y., Hiller, S., Wilder, J. Tang, C., Tang, L., Wu, X., Xiong, W., Ruan, H.,
& Reddick, R. (2000) Aortic wall damage in mice Hussain, M., Wu, J., Zou, C. & Wu,
unable to synthesize ascorbic acid. Proc Natl Acad
Sci U S A, 97, 841-846. X. (2016) Glioma-associated Oncogene 2 Is
Essential for Trophoblastic Fusion by Forming a
Moeschel, K., Beck, A., Weigert, C., Lammers, R., Transcriptional Complex with Glial Cell Missing-
Kalbacher, H., Voelter, W., Schleicher, E.D., a. J Biol Chem, 291, 5611-5622.
Haring, H.U. & Lehmann, R. (2004) Protein kinase Wang, X.A., Zhang, R., She, Z.G., Zhang, X.F.,
C-zeta-induced phosphorylation of Ser318 in Jiang, D.S., Wang, T., Gao, L., Deng,
insulin receptor substrate-1 (IRS-1) attenuates the
interaction with the insulin receptor and the W., Zhang, S.M., Zhu, L.H., Guo, S., Chen, K.,
tyrosine phosphorylation of IRS-1. J Biol Chem, Zhang, X.D., Liu, D.P. & Li, H. (2014) Interferon
279, 25157-25163. regulatory factor 3 constrains IKKbeta/NF-kappaB
Nakajima, K., Yamauchi, K., Shigematsu, S., Ikeo, S., signaling to alleviate hepatic steatosis and insulin
Komatsu, M., Aizawa, T. & Hashizume, K. (2000) resistance. Hepatology, 59, 870-885.
Selective attenuation of metabolic branch of Wang, F., Han, L. & Hu, D. (2017) Fasting insulin, insulin
insulin receptor down-signaling by high glucose in resistance and risk of hypertension in the general
a hepatoma cell line, HepG2 cells. J Biol Chem, population: A meta-analysis. Clin Chim Acta, 464,
275, 20880-20886. 57-63.
Nandipati, K.C., Subramanian, S. & Agrawal, D.K.
(2017) Protein kinases: mechanisms and Xu, H., Barnes, G.T., Yang, Q., Tan, G., Yang, D., Chou,
downstream targets in inflammation-mediated C.J., Sole, J., Nichols, A., Ross, J.S., Tartaglia,
obesity and insulin resistance. Mol Cell Biochem, L.A. & Chen, H. (2003) Chronic inflammation in
426, 27-45. fat plays a crucial role in the development of
Siman, C.M. & Eriksson, U.J. (1997) Vitamin C obesity-related insulin resistance. J Clin Invest,
supplementation of the maternal diet reduces the 112, 1821-1830.
rate of malformation in the offspring of diabetic Yu, C., Chen, Y., Cline, G.W., Zhang, D., Zong, H., Wang,
rats. Diabetologia, 40, 1416-1424. Y., Bergeron, R., Kim, J.K., Cushman, S.W.,
Sabio, G., Das, M., Mora, A., Zhang, Z., Jun, J.Y., Ko, Cooney, G.J., Atcheson, B., White, M.F., Kraegen,
H.J., Barrett, T., Kim, J.K. & Davis, R.J. (2008) A E.W. & Shulman, G.I. (2002) Mechanism by
stress signaling pathway in adipose tissue which fatty acids inhibit insulin
regulates hepatic insulin resistance. Science, 322, activation of insulin receptor substrate-1 (IRS-1)-
1539-1543. associated phosphatidylinositol 3-kinase activity in
Souza-Mello, V., Gregorio, B.M., Cardoso-de- muscle. J Biol Chem, 277, 50230-50236.
Lemos, F.S., de Carvalho, L., Aguila, Yang, Z., Mo, X., Gong, Q., Pan, Q., Yang, X.,
Cai, W., Li, C., Ma, J.X., He, Y. & Gao,
M.B. & Mandarim-de-Lacerda, C.A. (2010)
Comparative effects of telmisartan, sitagliptin and G. (2008) Critical effect of VEGF in the process of
metformin alone or in combination on obesity, endothelial cell apoptosis induced by high glucose.
insulin resistance, and liver and pancreas Apoptosis, 13, 1331-1343.
remodelling in C57BL/6 mice fed on a very high- Yao, H., Shi, W., Wu, J., Xu, C., Wang, J., Shao,
fat diet. Clin Sci (Lond), 119, 239-250. Y., Wu, X. & Zhang, Z. (2015)
Endothelial Rac1 is essential for hematogenous
metastasis to the lung. Oncotarget, 6, 17501-
17513.
Zang, M., Zuccollo, A., Hou, X., Nagata, D., Walsh, K.,
Herscovitz, H., Brecher, P., Ruderman, N.B. &
Cohen, R.A. (2004) AMP-activated protein kinase
is required for the lipid-lowering effect of
metformin in insulin-resistant human HepG2 cells.
J Biol Chem, 279, 47898-47905.
Zabolotny, J.M., Kim, Y.B., Welsh, L.A.,
Kershaw, E.E., Neel, B.G. & Kahn, B.B. (2008)
Protein-tyrosine phosphatase 1B expression is
induced by inflammation in vivo. J Biol Chem,
283, 14230-14241.
Zhao, H., Huang, X., Jiao, J., Zhang, H., Liu, J., Qin, W.,
Meng, X., Shen, T., Lin, Y., Chu, J. & Li, J. (2015)
Protein phosphatase 4 (PP4) functions as a critical
regulator in tumor necrosis factor (TNF)-alpha-
induced hepatic insulin resistance. Sci Rep, 5,
18093.
Figure legends: mean ± S.E.M (n=8). *P <0.05 vs. the LD group,
#P<0.05 vs. the TNF-α + LD group.
Figure 1 Vitamin C deficiency aggravated TNF- Figure 5 Vitamin C improved the TNF-α-
α-induced insulin resistance in Gulo-/- mice. (A) induced glucose and lipid metabolic disorder in
Mice from each group were starved for 14 h, HepG2 cells
subsequently intraperitoneally injected with 1 g/kg (A) Cell viability determined by cell counting kit-8
of glucose, and blood glucose concentrations were in HepG2 cells treated with vitamin C (0-200 μM)
measured at the indicated time points. (B) Blood for the indicated time (n=8). (B and C) Glycogen
glucose values for the area under the curve (AUC) levels determined by the glycogen assay kit in
were calculated during GTT. (C) Fasting serum HepG2 cells treated with vitamin C for the
insulin level. indicated time and dose, and or TNF-α (10 ng/ml,
(D) The HOMA-IR index was calculated 24 h; n=5). (D-G) HepG2 cells were cultured with
according to the formula [FBG (mmol/l) × Fins vitamin C (100 μM) and or TNF-α (10 ng/ml) for
(mIU/l)] / 22.5. (E) Hepatic glycogen level. (F) 24 h, and
Relative mRNA levels of PEPCK and G6P in liver 2-NBD glucose uptake were measured using a flow
tissues. Data were expressed as mean ± S.E.M cytometer (n=5) (D). The relative mRNA levels of
(n=8). *P <0.05 vs. the LD group, #P<0.05 vs. the genes were measured by qRT-PCR (n=5, E and F).
TNF-α + LD group. LD: 0.33 g/L of vitamin C; The protein expression of PEPCK, G6P, FAS, P-
HD: ACC and ACC were determined by western blot
3.3 g/L of vitamin C. (n=3, G). The representative western blot (left) and
quantification (right) are shown. Data were
Figure 2 Vitamin C deficiency aggravated TNF- expressed as mean ± S.E.M. *P <0.05 vs. the
α-triggered hepatic lipid accumulation in Gulo-/- Control. #P<0.05 vs. the TNF-α group. VC:
mice vitamin C.
(A) Hematoxylin and eosin (H&E) staining of Figure.6 Vitamin C restored TNF-α-negated
liver tissues (scale bar: 50 µm, magnification: GLUT2 levels and the IRS-1/AKT/GSK3β
400×). (B) Liver triglyceride content. (C) Liver pathway in HepG2 cells
cholesterol content. (D) Relative hepatic mRNA HepG2 cells were treated with vitamin C (100 μM)
levels of ACC1 and FAS in liver tissues. Data were and or TNF-α (10 ng/ml) for 24 h, and stimulated
expressed as mean ± S.E.M (n=8). *P <0.05 vs. with insulin (100 nM) for 10 min. The expression
the LD group, #P<0.05 vs. the TNF-α + LD group. and phosphorylation of IRS-1, AKT, GSK3β and
Figure 3 Vitamin C deficiency increased the GLUT2 were determined by western blot (A and
TNF-α-induced inactivation of the AKT/GSK3β B). The representative western blot (left) and
pathway in Gulo-/- mice quantification (right) are shown. Data were
expressed as mean ± S.E.M (n=3). *P <0.05 vs.
Liver tissue lysates from each group were the Control. #P<0.05 vs. the TNF-α group.
prepared and analyzed by western blot. Figure.7 Vitamin C inhibited the TNF-α-
Representative western blot analysis (left) and induced activation of the
quantification (right) of the expression and
phosphorylation levels of AKT and GSK3β in the IKKβ/IκBα/NF-κB pathway in HepG2 cells
liver homogenate of mice in each group (n=8). *P
<0.05 vs. the LD group, #P<0.05 vs. the TNF-α + HepG2 cells were treated with TNF-α (10 ng/ml)
LD group. for the indicated time, or treated with vitamin C
Figure 4 Vitamin C deficiency aggravated (100 μM) for 8 h, before cells were treated with
TNF-α-induced inflammation in Gulo-/- mice TNF-α (10 ng/ml) for 15 min. The time for the
activation of the IKKβ/IκB-α/NF-κB signaling
(A) Relative mRNA levels of IL-1α and IL-6 in liver pathway induced by TNF-α were determined by
tissues of Gulo-/- mice. (B) IL-1α and IL-6 levels western blot (A). The expression of
determined by ELISA in liver tissues of Gulo-/- mice.
(C). P-NF-κBp65 in the cytoplasm and nuclei were
NF-κBp65-positive expression (brown) in liver determined by western blot (B) and
tissues, as determined by immunohistochemistry immunofluorescence staining (C). The expression
(scale bar: 50 µm, magnification: 200×). and phosphorylation of IKKβ and IκBα were
(D).Western blot analysis (left) and quantification determined by western blot (D). The representative
(right) of NF-κBp65 expression in nuclei of the western blot (left) and quantification (right) are
liver homogenate of mice. Data were expressed as shown. Data were expressed as means ± S.E.M
(n=3). *P <0.05 vs. controls or 0 min. #P<0.05 vs.
the TNF-α or 30 min group.

Figure 8 Vitamin C inhibited the TNF-α-


mediated activation of JNK and p38 rather than
ERK in HepG2 cells
HepG2 cells were treated with TNF-α (10 ng/ml)
for the indicated time or treated with vitamin C
(100 μM) for 8 h before cells were treated with
TNF-α (10 ng/ml) for 30 min. The time for the
activation of MAPKs induced by TNF-α were
determined by western blot (A). The expression
and phosphorylation of JNK, p38 and ERK were
determined by western blot (B). The
representative Western blot (left) and
quantification (right) are shown. Data are expressed
as mean ± S.E.M (n=3). *P <0.05 vs. Controls or 0
min. #P<0.05 vs. the TNF-α or 60 min group.

Figure.1 Vitamin C up-regulated high glucose-


negated VEGF expression in HUVECs
(A) HUVECs were treated with vitamin C (100
μM) for 8 hours before the cells were exposed to
25 mM glucose, expression of VEGF was
determined by Western blot (n=3). Representative
Western blot (left) and quantification (right). (B)
VEGF level in culture supernatant was measured
by ELISA (n=5). Data are expressed as means ±
SEM. *P <0.05 versus Control. #P<0.05 versus
HG. Glucose concentration (5.5 mM) as a control
group, HG: 25 mM glucose.
Figure 1A
Figure 1B
Figure 1C
Figure 1D
Figure 1E
Figure 1F
Figure 2A
Figure 2B
Figure 2C
Figure 2D
Figure 3
statistics for Figure 3
Figure 4A
Figure 4B
Figure 4C
Figure 4D
statistics for Figure 4D
Figure 5A
Figure 5B
Figure 5C
Figure 5D
Figure 5E
Figure 5F
Figure 5G
statistics for Figure 5G
Figure 6A
Figure 6B
statistics for Figure 6B
Figure 7A
statistics for Figure 7A
Figure 7B
Figure 7C
Figure 7D
statistics for Figure 7B and 7D
Figure 8A
statistics for Figure 8A
Figure 8B
statistics for Figure 8B
Graphical Abstract

Anda mungkin juga menyukai