You are on page 1of 8

Biochemical and Biophysical Research Communications 476 (2016) 204e211

Contents lists available at ScienceDirect

Biochemical and Biophysical Research Communications


journal homepage: www.elsevier.com/locate/ybbrc

ROS-mediated glucose metabolic reprogram induces insulin resistance


in type 2 diabetes
Kelei Dong, Hua Ni, Meiling Wu, Ziqing Tang, Michael Halim, Dongyun Shi*
Department of Biochemistry and Molecular Biology, Shanghai Medical College of Fudan University, Free Radical Regulation and Application Research Center
of Fudan University, Shanghai 200032, Peoples Republic of China

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

Article history: Oxidative stress is known to contribute to insulin resistance in diabetes, however the mechanism is not
Received 11 May 2016 clear. Here we show that reactive oxygen species (ROS) could reprogram the glucose metabolism through
Accepted 16 May 2016 upregulating the pentose pathway so as to induce insulin resistance in type 2 diabetes (T2DM). By using
Available online 17 May 2016
streptozotocin-high fat diet (STZ-HFD) induced T2DM in rats, we show that diabetic rats exhibited high
level of oxidative stress accompanied with insulin resistance. Hypoxia inducible factor (HIF-1a) protein
Keywords:
expression as well as its downstream target glucokinase (GK), were upregulated; The glycogen synthesis
Reactive oxidative stress
increased accordingly; However the glycolysis was inhibited as indicated by decreased
Insulin resistance
Pentose pathway
phosphofructokinase-1 (PFK-1), pyruvate kinase (PK), phospho-PFK-2/PFK-2 (p-PFK-2/PFK-2) ratio,
Glycolysis lactate dehydrogenase (LDH) and pyruvate dehydrogenase kinase (PDK); Pyruvate dehydrogenase (PDH)
Type 2 diabetes mellitus which promotes pyruvate to generate acetyl-CoA declined as well. While phospho-acetyl-CoA carbox-
ylase/acetyl-CoA carboxylase (p-ACC/ACC) ratio increased, meaning that lipid beta-oxidation increased.
The pentose pathway was activated as indicated by increased G6PD activity and NADPH level. Our results
suggest that diabetic rats countervail ROS stress through increasing pentose pathway, and reprogram the
energy metabolic pathway from glycolysis into lipid oxidation in order to compensate the ATP
requirement of the body, which causes insulin resistance.
2016 Elsevier Inc. All rights reserved.

1. Introduction with vitamin E, vitamin C, or glutathione improves insulin sensi-


tivity in insulin-resistant individuals [4,5]. The recent nding that
In recent years, diabetes has been characterized as a global insulin resistance is associated in humans with reduced intracel-
epidemic, with numbers expected to reach 300 million by 2025 [1]. lular antioxidant defense also support that oxidative stress is
T2DM is considered to be attributable to peripheral insulin resis- involved in insulin resistance [6].
tance. Insulin resistance plays a pivotal role in the process of T2DM It is well known that excessive calorie intake, combined with
and it is the main pathological process of T2DM, however its little physical activity will lead to a situation of insulin resistance.
mechanisms are still unclear [2]. When caloric intake exceeds the energy expenditure, the substrate-
Oxidative stress is a phenomenon that arises when an imbal- induced increase in citric acid cycle activity generates an excess of
ance occurs to the production and elimination of ROS and it is mitochondrial NADH (mNADH) [7]. When excessive NADH cannot
contributed to the occurrence and development of T2DM. Oxidative be dissipated by oxidative phosphorylation (or other mechanisms),
stress has been shown to be related to insulin resistance. Some the mitochondrial proton gradient increases and single electrons
studies suggest that oxidative stress is responsible for altering are transferred to oxygen, leading to the formation of free radicals,
intracellular signaling pathways, which thereby inducing the pos- particularly superoxide anion [8]. To protect themselves against
sibility of insulin resistance [3]. In vitro studies and in animal ROS damage, cells would reduce ROS generation or increase ROS
models, antioxidants have been shown to improve insulin sensi- removal. By inhibiting the uptake of insulin stimulating nutrient
tivity [4]. Several clinical trials have demonstrated that treatment and preventing high energy compounds (pyruvate, fatty acids)
from entering into the mitochondria, mNADH accumulation is
hindered and this eventually reduced ROS generation. This may be
* Corresponding author. Tel.: 86 21 54237299; fax: 86 21 54237897. one of the reasons as to how ROS causes insulin resistance,
E-mail address: dyshi@fudan.edu.cn (D. Shi). nevertheless, the mechanism is still unclear.

http://dx.doi.org/10.1016/j.bbrc.2016.05.087
0006-291X/ 2016 Elsevier Inc. All rights reserved.
K. Dong et al. / Biochemical and Biophysical Research Communications 476 (2016) 204e211 205

It is known that cancer cells exhibit high basal levels of ROS measured according to the method as described in Ref. [17]. NADPH
owning to activation of oncogenes and tumor suppressor genes and oxidase (NOX) activity was measured as previously described [18].
tumor microenvironment deletions. However such substantial Enzyme activity was standardized using protein quantication.
amount of ROS will lead to oxidative stress. Studies have shown
that cancer cells have special ability to counter the oxidative stress
2.4. GSH/GSSG ratio measurement
by upregulating the pentose pathway [9]. Our recent study also
found that tumor cells can adaptively generate strong antioxidant
Livers were cut into 50 ml 1 M HPO3 for GSH detection. After 2e3
pathways against oxidative stress [10]. Based on the high level of
freeze/thaw cycles, the suspension was centrifuged with
oxidative stress in diabetes, we speculated that diabetes may also
12,000 rpm for 10 min at 4  C and then assayed as previously
possess a similar effect, which results in insulin resistance. In this
described [19].
study, we established STZ-HFD induced type 2 diabetic rat model,
we found that, similar with the cancer cells, diabetic rats exhibited
the upregulated HIF-1a, increased GK as well as enhanced the 2.5. NADH and NADPH assay
pentose pathway. Moreover, the diversion of glucose-6-phophate
(G6P) into the pentose pathway impaired glycolysis, thus repro- Cells were scraped into 200 ml cold 40 mM NaOH for NADH and
gram the energy metabolic pathway from glycolysis into lipid NADPH detection. After 2e3 freeze/thaw cycles, the suspension was
oxidation to compensate the ATP requirement of the body, there- centrifuged with 12,000 rpm for 10 min at 4  C and then assayed as
fore resulting insulin resistance. This new point will provide new previously described [20].
ideas for T2DM treatment.
2.6. Free fatty acids measurement
2. Material and methods
Serum and liver free fatty acids (FFA, nonesteried fatty acids)
2.1. Diabetic animal models were measured as previously described in Ref. [21].

All animal-related procedures were approved by the Fudan


University Institutional Laboratory Animal Ethics Committee. Male 2.7. Western blot
SD rats (150e160 g body weight) were obtained from Fudan Uni-
versity Animal Center (Shanghai, China). Induction of type 2 dia- Western blot was performed according to standard procedures
betes by STZ (Sigma Aldrich, USA)-high fat diet (STZ-HFD) was and protein extracts from liver was prepared as previously
performed as previously described [11,12]. Following a 12 h fasting described [22]. Protein expression was analyzed with antibodies
period, the rats were randomly divided into ve groups - i. control: against GK, PFK-1, PK, GYS-1, HIF-1a, LDH, PDH, PDK and GLUT4
Chow; ii. diabetes: HFD; iii. diabetes: HFD STZ (35 mg/kg); . (Proteintech, USA) and p-PFK-2, PFK-2, p-GYS, p-ACC, ACC and b-
diabetes: HFD STZ (50 mg/kg); . diabetes treated with apocynin actin (Cell Signaling Technology, USA). Protein expression is visu-
(Apo): HFD STZ (50 mg/kg) Apo; (n 8e10). The control group alized on Tanon-5200 Chemi-luminescent Imaging System (Tanon
rats were fed with a standard rodent chow diet whereas the others Science Technology).
received a single intraperitoneal (ip) injection of streptozotocin
(STZ, Sigma) in 0.1 M sodium citrate buffer solution (pH 4.5) and 2.8. MDA measurement
were fed with HFD diet. Apo was dissolved in the water. For the
HFD STZ (50 mg/kg) Apo groups, rats were pretreated with Apo The measurement of plasma malondialdehyde (MDA) experi-
for 30 min, and then injected with STZ (50 mg/kg BW). Apo was ment was performed according to the manufacturers instructions
administered on a daily basis at 30 mg/kg BW. of MDA kit (Nanjing Jiancheng Biological Engineering Institute,
Normal chow and high-fat diet (HFD) were purchased from Nanjing, China). The measurement principle is the usage of
Shanghai SLRC laboratory animal Company Ltd (Shanghai, China) degradation products of lipid peroxidation MDA with thiobarbituric
and the nutritional composition was shown in Supplementary acid (TBA) condensation to form a red product which has a
Table S1. maximum absorption peak at 532 nm, and can be measured by a
After 2 months of treatment, blood samples were drawn for colorimetric MDA content.
plasma and serum extraction. Following that, all rats were killed
and their livers were systematically removed and then frozen at
2.9. TAOC measurement
80  C for each index detection and also to perform further assays.

The measurement of plasma total antioxidant capacity (TAOC)


2.2. Oral glucose tolerance test (OGTT), insulin tolerance test (ITT)
experiment was performed according to the manufacturers in-
and insulin secretion
structions of TAOC kit (Nanjing Jiancheng Biological Engineering
Institute, Nanjing, China).
Measurement of OGTT was performed according to the proce-
dure previously described [13]. Alternatively, after a 12 h fasting
period, rats were administered with glucose of 2.5 g/kg body 2.10. Statistical analysis
weight. ITT was performed according to the procedure previously
described [14]. Accu-Chek Performa (Roche Diagnostics, Germany) Linear regression statistical analysis was performed using
was used to measure blood glucose levels in rats. Insulin secretion GraphPad Prism 5.0. Unpaired Student t-tests were used to calcu-
was detected according to the procedure previously described [15]. late the signicant difference between two sample groups. For
datasets containing more than two groups that required analysis of
2.3. Enzyme activities changes overtime, one-way ANOVA and Tukey post hoc test cal-
culations were performed instead. All data are expressed as
Liver GK activity was measured as described previously [16]. mean SD. P values of less than 0.05 were considered to be sta-
Glucose-6-phosphate dehydrogenase (G6PD) activity was tistically signicant between the given data sets.
206 K. Dong et al. / Biochemical and Biophysical Research Communications 476 (2016) 204e211

3. Results level declined in diabetic rats (Fig. 2C), suggesting these diabetic
rats were underwent oxidative stress. Furthermore, NADH content
3.1. HFD combined with STZ treatment induces insulin resistance in also increased signicantly in diabetic rats (Fig. 2D), which impli-
diabetic rats cates impaired oxidation phosphorylation and mitochondrial ROS
generation. The expression of glucose transporter GLUT4 decreased
In our experiment, we induced type 2 diabetic rats by combining in diabetic rats (Fig. 2E). Interestingly, although diabetic rats
STZ and HFD treatment. In HFD treated group, fasting and post- exhibited oxidative stress, GSH/GSSG ratio did not show signicant
prandial blood glucose did not alter signicantly (Fig. 1A), but in- change (Fig. 2F). We found that G6PD increased signicantly in
sulin levels are signicantly increased (Fig. 1B), hence showing a those diabetic rats (Fig. 2G). This suggests that pentose phosphate
marked hyperinsulinemia situation, nonetheless overt diabetes are pathway was activated in response to oxidative stress. NADPH
not manifested. In HFD-STZ combination treated rats, the glucose increased in parallel (Fig. 2H), presumably to maintain the GSH
tolerance was impaired in a STZ dose-dependent manner (Fig. 1C). levels which can protect against ROS stress. However, the ratio of
In the 35 mg/kg STZ group, although fasting blood glucose did not NADPH/NADP was declined (Fig. 2H). We supposed that NADPH
show signicant increase, postprandial blood glucose increased was consumed to offer reducing equivalents to GSSG, and NADPH
signicantly (Fig. 1D). Meanwhile insulin tolerance test also itself being converted to NADP, thus caused the declined NADPH/
showed that insulin resistance may occur in diabetic rats (Fig. 1E). NADP ratio. Our results suggest that enhanced PPP generates
With increasing doses of STZ, blood glucose increased in both fas- NADPH to countervail oxidative stress in diabetic rats.
ted and fed condition. 50 mg/kg STZ treated group exhibited overt
diabetes (Fig. 1D). 3.3. Enhanced G6PD diverts G6P into pentose pathway from
glycolysis
3.2. Oxidative stress in diabetic rats compensatorily enhanced
pentose pathway It is known that HIF-1a is an important transcription factor in
regulating glycolysis. Our previous study showed that Hif-1a could
Our results show that MDA level increased and NOX activity be stabilized by hypoxia or ROS in tumor [23]. We speculated Hif-1a
which causes cytosolic ROS, was activated (Fig. 2A-B), while TAOC might also be upregulated in diabetic rats which were underwent

Fig. 1. HFD combined with STZ treatment induces insulin resistance in diabetic rats. AeB: Wild-type male SD rats were fed with HFD for 2 months and then blood glucose and
plasma insulin level were measured. A: Fasting and postprandial blood glucose levels of Chow and HFD groups. B: Fasting and postprandial plasma insulin levels of Chow and HFD
groups. C-E: Wild-type male SD rats were fed with Chow or HFD STZ (35 and 50 mg/kg, respectively) for 2 months and then killed for further analysis. C: OGTT. D: Fasting and
postprandial blood glucose levels in Chow and HFD STZ groups. E: ITT. n 8e10 per group. *P < 0.05, **P < 0.01, ***P < 0.001 compared with Chow group.
K. Dong et al. / Biochemical and Biophysical Research Communications 476 (2016) 204e211 207

Fig. 2. Oxidative stress in diabetic rats compensatorily enhanced pentose pathway. A: Measurement of plasma MDA level in rats. B: NOX activity analysis in rat livers. C: Mea-
surement of plasma TAOC level in rats. D: NADH content analysis. E: Protein expression of GLUT4 in rat livers. F: GSH/GSSG ratio analysis in rat livers. G: G6PD activity analysis in rat
livers. H: Measurement of NADPH and NADPH/NADP ratio in rat livers. n 8e10 per group. *P < 0.05, **P < 0.01, ***P < 0.001 compared with Chow group.

oxidative stress. Our results showed that HIF-1a expression was (Fig. 3C). Moreover, PDH which catalyze pyruvate to form acetyl-
signicantly increased in diabetic rat livers (Fig. 3A). Its down- CoA was also inhibited (Fig. 3C). We also analyze the synthesis of
stream target GK (HK), the key enzymes in the glycolytic pathway, glycogen. As shown in Fig. 3B, the p-GYS-1/GYS-1 decreased which
was also upregulated in diabetic rats (Fig. 3A). Increased GK activity indicates the increased glycogen synthesis. These results suggest
could produce more gluocose-6-phosphate (G6P) and should in- that the ux of G6P into PPP and glycogen synthesis enhanced,
crease glycolysis. Interestingly, the glycolysis was inhibited. We however, the ux of G6P into the glycolysis was inhibited. Therefore
found that the expression of PFK-1 PK, LDH and PDK, the enzymes the energy generation derived from glucose oxidation impaired in
in the glycolytic pathway, was signicantly lower in diabetic rats diabetes.
(Fig. 3C). The phospho-PFK-2 is the bifunctional enzyme that syn-
thesizes fructose 2,6-bisphosphate (F2,6BP), the allosteric regulator 3.4. ROS reprogram energy pathway into lipid oxidation
of PFK-1. We found that phospho-PFK-2/PFK-2 (p-PFK-2/PFK-2)
ratio which implicates the glycolytic rates signicantly decreased Since ROS impaired energy generation derived from glucose, we
208 K. Dong et al. / Biochemical and Biophysical Research Communications 476 (2016) 204e211

Fig. 3. Enhanced G6PD diverts G6P into pentose pathway from glycolysis. A: Protein expression of HIF-1a and GK and GK activity analysis in rat livers. B: Protein expression of p-
GYS in rat livers. C: Protein expression of PFK-1, PK, p-PFK-2, LDH, PDH, PDK in rat livers. n 8e10 per group. *P < 0.05, **P < 0.01, ***P < 0.001 compared with Chow group.

speculate the other pathway should be upregulated in order to 4. Discussion


meet the ATP requirement in diabetic rats. We found p-ACC/ACC
ratio increased which means lipid beta-oxidation in diabetic rats Insulin resistance is an important decisive factor in T2DM
increased (Fig. 4A). The FFA levels were signicantly increased metabolic abnormalities. Oxidative stress plays an important role in
(Fig. 4B). After inhibition cellular ROS by Apo treatment, FFA levels inducing insulin resistance, but the mechanism behind it remains
in diabetic rats decreased signicantly (Fig. 4C). This suggests ROS unclear.
involved in reprograming glycolysis into lipid oxidation. Apo In our experiment, we established the model of T2DM by HFD-
treatment also improved GLUT4 expression and postprandial blood STZ. Studies shown that prior to insulin resistance, the body may
glucose (Fig. 4D and E), suggesting that ROS induced insulin resis- exhibit at near normal blood glucose level owing to hyper-
tance could be alleviated by antioxidant intervention. insulinemia. Our studies revealed that in HFD group, fasting and
K. Dong et al. / Biochemical and Biophysical Research Communications 476 (2016) 204e211 209

Fig. 4. ROS reprogram energy pathway into lipid oxidation. A: Protein expression of p-ACC in rat livers. B: Measurement of liver and serum FFA levels in diabetic rats. C: Mea-
surement of liver and serum FFA levels in Apo intervention rats. D: Protein expression of GLUT4 in Apo intervention rat livers. E: Postprandial blood glucose levels in Apo
intervention rats. *P < 0.05, **P < 0.01, ***P < 0.001 compared with Chow group except in (C-E, HFD STZ (50 mg/kg) group).

postprandial blood glucose did not change signicantly, however, occurs, this causes a lot of substrate to enter the citric acid cycle,
insulin levels became signicantly higher, herein showing a which in turn leads to an excessive mitochondrial acetyl-CoA and
marked hyperinsulinemia situation, but overt diabetes did not NADH production [7]. Acetyl-CoA, derived either from glucose
appear. We know that STZ is widely used to induce experimental through pyruvate or from beta-oxidation of FFA, combines with
diabetes in animals. It was found to generate reactive oxygen spe- oxaloacetate to form citrate, which enters the citric acid cycle and is
cies and reactive nitrogen. STZ enters the beta cell via a glucose converted to isocitrate. NAD-dependent isocitrate dehydrogenase
transporter (GLUT2) and causes the formation of superoxide radi- generates NADH. The accumulation NADH could cause ROS gener-
cals. Consequently, hydrogen peroxide and hydroxyl radicals are ation [7]. The persistence of such an imbalance between caloric
also generated [24]. Essentially, STZ-induced oxidative stress occurs intake and expenditure can lead to accumulation of ROS, which can
in the incidence of T2DM. In HFD-STZ group we show that as STZ consequently lead to overt diabetes. Therefore diabetes caused by
dose increases, glucose tolerance decreases in diabetic rats. In the obesity generally required a long period of time. In our model, by
35 mg/kg STZ group, although there is no signicant increase in increasing the oxidative stress, it can promote the accumulation of
fasting blood glucose, postprandial blood glucose increased ROS to a certain threshold, which can in turn promote overt dia-
signicantly. Meanwhile insulin tolerance test also showed that betes occurring ahead of time.
insulin resistance may occur in diabetic rats. When islet b cells are According to our study, in addition to the signicant increase in
no longer able to through increasing insulin compensate for insulin oxidative stress in diabetic rats, NADH content also increased
resistance, impaired glucose tolerance appeared. With increasing signicantly, especially in the 50 mg/kg STZ group. Mitochondrial
doses of STZ, 50 mg/kg STZ group showed overt diabetes, where ROS could activate PKC and NF-kB and favor the overexpression of
hyperglycemia is apparent. Our experiments show that in diabetic NOX, which could further generate a great amount of superoxide.
rats, the expression of GLUT4 is signicantly decreased, which Our results show that NOX activity increased, which resulted in a
further proves that insulin resistance occurs in diabetic rats. Insulin vicious cycle of ROS in vivo, thus leading to rising levels of MDA and
resistance leads to a reduction in the amount of glucose that is declining levels of TAOC. ROS can damage DNA. DNA damage is an
transported into the muscle cells, and lipolysis increased, which obligatory stimulus for the activation of the nuclear enzyme poly
explains why the FFA levels are signicantly increased. (ADP-ribose) polymerase, resulting in reduced GLUT4 expression
In the HFD model, owning to high fat diet intake, energy surplus and the subsequent insulin resistance. In our experiments, GLUT4
210 K. Dong et al. / Biochemical and Biophysical Research Communications 476 (2016) 204e211

increased after being subjected to Apo intervention accompanied relieving oxidative stress may not be a wise decision because it may
by improvement in postprandial blood glucose, these results sug- increase the damage attributable to reactive oxygen species.
gest that oxidative stress could through impairing GLUT4 lead to
insulin resistance. Author contributions
HIF-1a is a hypoxia-inducible factor, rst discovered in
mammalian cells, where oxygen concentration is low. Recent K.D. carried out the major experiments; H.N. assisted with the
studies reveal that HIF-1a expression does not only occur in the establishment of diabetic animal models; M.W. carried out the
absence of oxygen, oxidative stress can also increase the expression HPLC measurement; Z.T carried out the GSH/GSSG ratio measure-
of HIF-1a [23]. Owning to the fact that T2DM exhibits high levels of ment; M.H. assisted with the western blot and proof reading; D.S.
oxidative stress, we speculate that there is a high expression of HIF- designed and supervised experiments and wrote the manuscript.
1a. Our results show that diabetic rats had oxidative stress micro-
environment and increased HIF-1a level. The expression of HIF-1a Funding
enhanced with increasing concentration of STZ. We then analyzed
the downstream targets of HIF-1a. We found that GK which cata- This work was supported by National Natural Science Founda-
lyzes glucose into G6P was signicantly activated. G6P can ow into tion of China (31270901, 30970684).
glycolysis, PPP pathway and glycogen synthesis, respectively.
Interestingly, other enzymes in the glycolytic pathway such as PFK- Author disclosure statement
1, PK, p-PFK-2/PFK-2, PDK and LDH were all inhibited. PDH which
catalyzes pyruvate to form acetyl-CoA decreased, this indicates that No competing nancial interests exist for any of the authors.
oxidation phosphorylation derived from glucose was inhibited. Our
results suggest that both of aerobic and anaerobic respirations Acknowledgements
mediated by glycolysis are impaired in diabetic rats.
It is known that the upregulated PPP pathway could through The authors are sincerely grateful to Dr. Xiaodong Zhang from
increasing of NADPH enhance cellular oxidative buffering capacity Vero Biotech for his proof reading.
in response oxidative stress in tumor, i.e., a phenomenon existed in
the tumor Warburg effect [25]. Here we found, in diabetic rats, liver
Transparency document
cells also use this mechanism to reduce oxidative stress, PPP
pathway obviously enhanced. However, unlike the Warburg effect
Transparency document related to this article can be found
which has enhanced glycolysis, in diabetic rats, the upregulated PPP
online at http://dx.doi.org/10.1016/j.bbrc.2016.05.087.
pathway caused glycolytic substrates to be steered into the pentose
pathway, thereby leading to the declined glycolysis. Our results
Appendix A. Supplementary data
show that glycogen synthesis also enhanced in the diabetic rats.
These results suggest that majority G6P ows into glycogen syn-
Supplementary data related to this article can be found at http://
thesis and PPP pathway rather than glycolysis. We propose ROS
dx.doi.org/10.1016/j.bbrc.2016.05.087.
induced upregulation of G6PD diverts G6P into pentose pathway
from glycolysis, causing the declined glycolysis and the emergence
References
of insulin resistance.
Glycolysis is the foundation of both aerobic and anaerobic [1] M. Prentki, C.J. Nolan, Islet beta cell failure in type 2 diabetes, J. Clin. Invest.
respiration. The impaired glycolysis will lead to energy shortage. 116 (2006) 1802e1812.
Here we show that lipid oxidation was highly activated to [2] C.R. Kahn, Insulin resistance, insulin insensitivity, and insulin unresponsive-
ness: a necessary distinction, Metabolism 27 (1978) 1893e1902.
compensate to energy requirement of the body. Accordingly, the [3] J.L. Evans, I.D. Goldne, B.A. Maddux, et al., Are oxidative stress-activated
FFA level signicantly increased in liver cells and serum of diabetic signaling pathways mediators of insulin resistance and beta-cell dysfunc-
rats. Inhibition of ROS by Apo interference obviously decreased FFA tion? Diabetes 52 (2003) 1e8.
[4] G. Paolisso, D. Giugliano, Oxidative stress and insulin action: is there a rela-
level. It suggests that ROS is involved in reprograming energy
tionship? Diabetologia 39 (1996) 357e363.
pathway into lipid oxidation, which may contribute to the insulin [5] A. Ceriello, Oxidative stress and glycemic regulation, Metabolism 49 (2000)
resistance. 27e29.
Our results suggest that intracellular redox systems and cellular [6] C.R. Bruce, A.L. Carey, J.A. Hawley, et al., Intramuscular heat shock protein 72
and heme oxygenase-1 mRNA are reduced in patients with type 2 diabetes:
glucose metabolism are likely to inuence each other as a balanced evidence that insulin resistance is associated with a disturbed antioxidant
network. Upregulated PPP pathway is the compensatory effect of defense mechanism, Diabetes 52 (2003) 2338e2345.
the glucoses metabolism towards oxidative stress, and it may be [7] B.A. Maddux, W. See, J.J. Lawrence, et al., Protection against oxidative stress-
induced insulin resistance in rat L6 muscle cells by micromolar concentra-
closely related to many chronic diseases. Mechanism of insulin tions of alpha-lipoic acid, Diabetes 50 (2001) 404e410.
resistance is currently not very clear, this study discloses an [8] P. Maechler, L. Jornot, C.B. Wollheim, Hydrogen peroxide alters mitochondrial
important mechanism for insulin resistance: ROS, through activation and insulin secretion in pancreatic beta cells, J. Biol. Chem. 274
(1999) 27905e27913.
enhancing PPP pathway, reprograms the energy metabolic pathway [9] R.B. Hamanaka, N.S. Chandel, Cell biology. Warburg effect and redox balance,
from glycolysis into lipid oxidation to produce large amount of FFA, Science 334 (2011) 1219e1220.
causing NADH accumulation and ROS generation, which lead to [10] P. Li, D. Zhang, L. Shen, et al., Redox homeostasis protects mitochondria
through accelerating ROS conversion to enhance hypoxia resistance in cancer
GLUT4 downregulation. All factors such as impaired glycolysis, cells, Sci. Rep. 6 (2016) 22831.
decreased GLUT4 and increased FFA contribute to insulin [11] M.J. Reed, K. Meszaros, L.J. Entes, et al., A new rat model of type 2 diabetes: the
resistance. fat-fed, streptozotocin-treated rat, Metabolism 49 (2000) 1390e1394.
[12] K. Srinivasan, B. Viswanad, L. Asrat, et al., Combination of high-fat diet-fed and
Our results disclose that diabetic rats countervail ROS stress, and
low-dose streptozotocin-treated rat: a model for type 2 diabetes and phar-
reprogram the energy metabolic pathway from glycolysis into lipid macological screening, Pharmacol. Res. 52 (2005) 313e320.
oxidation to compensate the ATP requirement of the body, which [13] X. Wang, C. Gu, W. He, et al., Glucose oxidase induces insulin resistance via
causes insulin resistance. This may provide new ideas and ap- inuencing multiple targets in vitro and in vivo: the central role of oxidative
stress, Biochimie 94 (2012) 1705e1717.
proaches to T2DM treatment: relieving oxidative stress can alle- [14] S. Chen, W. Zhang, C. Tang, et al., Vanin-1 is a key activator for hepatic
viate insulin resistance, but inhibiting insulin resistance before gluconeogenesis, Diabetes 63 (2014) 2073e2085.
K. Dong et al. / Biochemical and Biophysical Research Communications 476 (2016) 204e211 211

[15] C. Wang, Y. Chi, J. Li, et al., FAM3A activates PI3K p110alpha/Akt signaling to dinucleotide, Anal. Biochem. 53 (1973) 452e458.
ameliorate hepatic gluconeogenesis and lipogenesis, Hepatology 59 (2014) [21] J.T. Rodgers, P. Puigserver, Fasting-dependent glucose and lipid metabolic
1779e1790. response through hepatic sirtuin 1, Proc. Natl. Acad. Sci. U. S. A. 104 (2007)
[16] S.D. Da, P. Ausina, E.M. Alencar, et al., Metformin reverses hexokinase and 12861e12866.
phosphofructokinase downregulation and intracellular distribution in the [22] D.M. Erion, S. Yonemitsu, Y. Nie, et al., SirT1 knockdown in liver decreases
heart of diabetic mice, IUBMB Life 64 (2012) 766e774. basal hepatic glucose production and increases hepatic insulin responsiveness
[17] M. Lamas-Maceiras, E. Rodriguez-Belmonte, M. Becerra, et al., KlGcr1 controls in diabetic rats, Proc. Natl. Acad. Sci. U. S. A. 106 (2009) 11288e11293.
glucose-6-phosphate dehydrogenase activity and responses to HO, cadmium [23] D.Y. Shi, F.Z. Xie, C. Zhai, et al., The role of cellular oxidative stress in regu-
and arsenate in Kluyveromyces lactis, Fungal Genet. Biol. 82 (2015) 95e103. lating glycolysis energy metabolism in hepatoma cells, Mol. Cancer 8 (2009)
[18] M.C. Castro, F. Francini, G. Schinella, et al., Apocynin administration prevents 32.
the changes induced by a fructose-rich diet on rat liver metabolism and the [24] T. Szkudelski, The mechanism of alloxan and streptozotocin action in B cells of
antioxidant system, Clin. Sci. (Lond) 123 (2012) 681e692. the rat pancreas, Physiol. Res. Acad. Sci. Bohemoslov. 50 (2001) 537e546.
[19] P.J. Hissin, R. Hilf, A uorometric method for determination of oxidized and [25] D. Anastasiou, G. Poulogiannis, J.M. Asara, et al., Inhibition of Pyruvate Kinase
reduced glutathione in tissues, Anal. Biochem. 74 (1976) 214e226. M2 by reactive oxygen species contributes to cellular antioxidant responses,
[20] C. Bernofsky, M. Swan, An improved cycling assay for nicotinamide adenine Science 334 (2011) 1278e1283.

You might also like