Skip to content
BY-NC-ND 4.0 license Open Access Published by De Gruyter Open Access December 29, 2017

Curcumin attenuates oxidative stress in liver in Type 1 diabetic rats

  • Zhenglu Xie EMAIL logo , Xinqi Zeng , Xiaqing Li , Binbin Wu , Guozhi Shen , Qianying Wu and Changbiao Wu
From the journal Open Life Sciences

Abstract

We investigated the effect of curcumin on liver anti-oxidative stress in the type 1 diabetic rat model induced by streptozotocin (STZ). Experimental diabetic rats were induced by STZ intraperitoneally. All rats were fed for 21 days including three groups of control (NC), diabetic model (DC) and curcumin-treated (Cur, 1.5 g/kg by gavage). The results showed that curcumin-treatment significantly decreased the blood glucose and plasma malondialdehyde levels, but significantly increased the plasma superoxide dismutase, glutathione peroxidase and reduced glutathione levels. Curcumin treatment decreased the activity of aldose reductase, but increased the plasma glucose-6-phosphate dehydrogenase, glucose synthetase and glucose-polymerizing activities. Curcumin treatment significantly decreased the protein of protein kinase C (PKC) and poly ADP ribose polymerase (PARP) expression in the Cur group compared with the DC group. Moreover, the sorbitol dehydrogenase activity was significantly decreased and deterred glucose enters into the polyol pathway leading to an increased NADPH content in the Cur group compared with the DC group. Our data provides evidence that oxidative stress in diabetic rats may be attenuated by curcumin by inhibiting polyol pathway associated with down-regulated expression of PKC and PARP, as evidenced by both an increase the antioxidant enzymes levels and glycogen biosynthesis enzymes activities.

1 Introduction

Diabetes mellitus (DM) is one of the most common endocrine metabolic disorders, which leads to glucose toxicity by increasing protein glycation and activating the polyol pathway associated with the protein of poly ADP ribose polymerase (PARP) and protein kinase C expression [1,2,3,4,5]. Normally, DM can generate a lot of reactive oxygen species (ROS) which can cause oxidative stress [6,7,8,9], resulting in inflammation, cell death and system organ dysfunction [10,11]. Previous studies have shown that the redox balance is disrupted by activating the polyol pathway and PARP [5,12]. Firstly, the glucose was transformed to fructose using NADH as coenzyme in the polyol pathway, leading to NADPH converting to NADH and over-production of the consumption of glucose [13,14]. Secondly, NADH was dehydrogenated to produce NAD+, which was used as a substrate to over-activated PARP resulting in the DNA oxidative damage in diabetes [15]. Therefore, restored redox balance would decrease levels of NADH and maybe alleviate the oxidative stress [16]. In addition, cells defend themselves by superoxide dismutase (SOD), glutathione peroxidase (GPx) and reduced glutathione (GSH) to combat ROS damage [17,18]. There is evidence that elevated plasma SOD levels can decrease the damage in diabetics [19,20]. Additionally, GPx can catalyze lipid hydroperoxides to lipid alcohols in a reaction that utilizes GSH as a co-substrate to reduce the effects of oxygen derived free radicals [21].

Curcumin is extracted from the rhizome of turmeric and has been shown to have many bioactivities involved in antioxidant, anti-inflammatory and metabolic disorders in different biological systems and animal models [22,23,24,25,26,27]. Given its beneficial effects, safety and cost-effectiveness, curcumin could be used in diabetes and its resulting complications as well as for obesity-related metabolic syndrome [9,28]. Moreover, previous studies have reported that curcumin can resist oxidative stress in the liver of high-fat diet (HFD)-induced diabetic rat by increasing oxygen consumption and the activity of SOD, GSH and GPx [25,29,30]. Based on the cellular redox balance, the aim of this study was to investigate the capacity of curcumin to attenuate the oxidative stress in STZ-induced diabetic rats by inhibiting the polyol pathway associated with the protein of PARP and PKC expression, as evidenced by both an increase in plasma antioxidant enzyme levels and glycogen biosynthesis enzyme activity within the liver.

2 Materials and methods

2.1 Chemicals

Streptozotocin (STZ) and curcumin were obtained from Sigma Chemicals Company (USA). The blood glucose, malondialdehyde (MDA), superoxide dismutase (SOD), sorbitol dehydrogenase (SDH), glutathione peroxidase (GPx) and reduced glutathione (GSH) kits were purchased from Nanjing Jiancheng Biologic, Inc. (Nanjing, China). Glucagon radioimmunoassay kit was procured from North Institute of Biological Technology (Beijing, China). The aldose reductase (AR), glucose-6-phosphate dehydrogenase (G6PD), glycogen synthetase (GS) and glucose-polymerizing enzyme (GPE) kits were purchased from BioAssay (Hayward, CA). Amplite colorimetric total NADP and NADPH assay kit was purchased from AAT Bioquest, Inc. (Sunnyvale, CA). All the other chemicals were of analytical grade.

2.2 Animals and ethics statement

Forty-five seven-week-old male Sprague-Dawley rats were obtained from Model Animal Research Center of Nanjing University (Nanjing, China). All rats were acclimatized to laboratory conditions, which included temperature (22±2 °C) and a 12 h light/dark cycle and were maintained on standard food pellets and tap water for one-week.

Ethical approval

All animal experimental procedures were approved by Fujian Agriculture and Forestry University Animal Care and Use Committee and the law of laboratory animals of the Fujian province Zoological Society.

2.3 Experimental design

All rats were weighted and randomly assigned; group I (NC, n=15) was a control group, group II (DC, n=15) was a diabetes control group, and group III (Cur, n=15) was a treated group. STZ in fresh 0.1 mol/L citrate buffer (pH 4.5) was administered intraperitoneally at a single dose of 80 mg/kg to establish diabetic rat model [31]. The control rats were also intraperitoneally administered with the citrate buffer. After two days, fasting blood glucose was tested from the tail vein of rats. Rats with diabetes were confirmed after fasting blood glucose levels exceeded 11.1 mmol/L. The curcumin treated group was put on a normal diet plus 1.5 g curcumin/kg body weight administered orally by intragastric intubation daily for 21 days [31,32,33].

2.4 Sampling

At the end of the experiment, animals were left to fast overnight and sacrificed by cervical decapitation after administering ether anesthesia. Blood was collected in heparin sodium vessels. Plasma was collected by centrifugation at 3500 rpm for 15 min and glucose concentration, MDA, SOD, SDH, GPx and GSH activities were measured. The liver was immediately collected and cleared of the adhering fat, weighed and homogenized, and then stored at -80°C until analysis.

2.5 Analytical methods

Blood glucose levels and plasma concentrations of MDA, SOD, SDH, GSH and GPx were determined using chemical kits according to the manufacturer’s instructions. The activities of SDH, GSH and GPx in the Liver were measured from homogenates. Liver tissue homogenates and hemolysates were prepared in Tris-HCl buffer (0.4 mol/L; pH 7.0). When testing the liver SDH, an aliquot of liver homogenate was treated with 1.6 mL triethanolamine buffer (0.2 mol/L; pH 7.4) and 0.2 mL NADH (12 mmol/L).

Glucagon in the liver was measured using a radioimmunoassay kit according to the manufacturer’s instructions. Samples, standards and controls were incubated with 125I-Glu and rabbit anti-Glu antibody for 24 h, and then they were separated by centrifugation. A separating medium was removed and the sediment was saved for measurement on a γ-counter. The activities of AR, G6PD, GPE and GS in the liver were measured spectrophotometrically using chemical kits according to the manufacturer’s instruction. A decrease of NADPH’s absorption or monitoring NADPH production at 340 nm was used to analyze AR activity and G6PD activity, based on coupling its production of glucose-6-phosphate to NADPH production.

2.6 SDS-PAGE and western blot analysis

Protein was extracted from frozen liver tissue. Protein concentration was measured using bovine serum albumin as the standard in the Bradford assay and 50 μg protein samples were heat denatured. Subsequently, the SDS-PAGE (10% gel) was performed to separate proteins according to standard procedures [34]. The protein antibody of anti-poly ADP ribose polymerase antibody (PARP, 1:1500, Abcam) and anti-protein kinase C antibody (PKC, 1:1500, Abcam) was used. The actin (1:1000, Abcam) was used as an internal control. Finally, the blot was washed and detected by enhanced chemiluminescence (ECL) using the LumiGlo substrate (Pierce, USA) and Clarity Western ECL Substrate (BioRad, USA).

2.7 Statistical analysis

The results were evaluated by one-way analysis of variance using the SPSS-20.0 software followed by LSD. Data was expressed as mean ± SE and statistical significance was considered when P<0.05.

3 Results

3.1 Blood glucose, plasma SOD and MDA concentration

As shown in Table 1, after 21 days of experiment, the blood glucose level was significantly increased in the DC group (P<0.001) and Cur group (P<0.05) compared with the NC group. However, curcumin-treated diabetic rats had significantly reduced blood glucose levels (P<0.001). Compared to the NC group, the level of MDA was significantly increased in the DC group (P<0.001), whereas this change was attenuated in the curcumin-treated diabetic rats (P<0.01). Furthermore, an obvious reduction of SOD activity in the DC group compared with the NC group (P<0.01), but in the Cur the SOD activity was increased significantly compared with that in the NC group (P<0.05) and DC group (P<0.001).

Table 1

Blood glucose, plasma levels of MDA and SOD

ItemGlucose (mmol/L)MDA (nmol/mL)SOD (U/mL)
NC[1]6.01±2.476.03±1.0325.01±3.47
DC[2]17.99±1.31 [***]9.22±0.67 [***]18.76±2.45 [**]
Cur[3]8.98±2.36 [*]. [###]6.38±0.24 [##]28.66±1.09 [*]. [###]

Note: the results are from an analysis of eighteen rats in each group. Data is presented as Means ± SE, indicates,

3.2 Hepatic glycogen content, activities of SDH, GPx and GSH in the liver and plasma

As shown in Table 2, the level of hepatic glycogen in the DC group was lower than that in the NC group (P<0.01), but curcumin treatment significantly increased the hepatic glycogen levels compared with the DC group (P<0.01). Data in Table 2 also includes the concentrations of SDH, GPx and GSH activities in the liver and plasma of all animals. The SDH activity in the DC group and Cur group was significantly higher (P<0.01) than that in the NC group. Curcumin treatment significantly decreased the activity of SDH in both the liver and plasma (P<0.01) compared with those in the DC group. The values of GPx and GSH were obviously decreased in both the liver (P<0.001, P<0.001, respectively) and plasma (P<0.01 and P<0.05, respectively) in the DC group compared with the NC group. Furthermore, curcumin treatment significantly increased the levels of GPx and GSH in the liver (P<0.001 and P<0.001, respectively) and plasma (P<0.01 and P<0.001, respectively) compared with those in the DC group.

Table 2

Blood glucose, plasma levels of MDA and SOD

ItemHG (mg/g)Liver SDH (Unit/g)Plasma SDH (Unit/mL)Liver GPx (μg glutathione / min●mg)Plasma GPx (μg glutathione / min●mg)Liver GSH (μg/mg)Plasma GSH (mg/dL)
NC[1]11.22±1.824.05±0.183.86±0.0811.75±0.1814.98±0.6823.61±1.1822.86±1.39
DC[2]5.62±1.26[**]8.28±0.43[***]5.55±0.74[*]6.58±0.13[***]10.35±0.23[**]10.72±0.43[***]13.55±0.91[*]
Cur[3]7.24±1.07[**][##]5.23±0.17[**][###]4.01±0.08[*], [##]11.23±1.23[###]14.35±0.71[##]21.07±0.81[*]. [###]20.77±0.21[*]. [###]

Note: the results are from an analysis of eighteen rats in each group. Data is presented as Means ± SE, indicates,

3.3 Redox imbalance parameters associated with liver enzyme activity

As results in Figure 1A demonstrate, the activity of aldose reductase was noticeably elevated in the DC group, but this change was inhibited in the Cur group. The enzyme systems convert glucose to fructose and NADPH to NADH, resulting in the significant decrease of NADPH (P<0.001) in the DC group when compared to the NC group (Figure 1B). In addition, NADPH content was lower, so GSH content was decreased as well. The activity of G6PD in the DC group was lower than in the NC group (Figure 1C), and the results suggested that decreased levels of NADPH could be partly driven by decrease of the G6PD activity. As shown in Figure 1D and 1E, the activities of glucose-polymerizing enzyme and glucose synthetase were reduced significantly in the DC group compared with the NC group (P<0.01, P<0.001, respectively). The curcumin treated diabetic rats notably increased the activities of glucose-polymerizing enzyme and glucose synthetase (P<0.05, P<0.01, respectively), but they were still lower than in the NC group.

Figure 1 The redox imbalance parameters associated with liver enzyme activity. A, aldose reductase activity; B, NADPH content; C, glucose-6-phosphate dehydrogenase (G6PD) activity; D, glucose-polymerizing activity; E, glucose synthetase activity. Data is presented as Means ± SE, indicates, * P<0.05, ** P<0.01 and *** P<0.001 means differences compared with NC group; # P<0.05, ## P<0.01, ### P<0.001 means differences compared with DC group.
Figure 1

The redox imbalance parameters associated with liver enzyme activity. A, aldose reductase activity; B, NADPH content; C, glucose-6-phosphate dehydrogenase (G6PD) activity; D, glucose-polymerizing activity; E, glucose synthetase activity. Data is presented as Means ± SE, indicates, * P<0.05, ** P<0.01 and *** P<0.001 means differences compared with NC group; # P<0.05, ## P<0.01, ### P<0.001 means differences compared with DC group.

3.4 The protein expression of the PKC and PARP

As shown in Figure 2A and 2B, the protein contents of PKC and PARP were notably increased in the DC group compared with the NC group (P<0.01, P<0.001, respectively). The protein expression of PKC and PARP was significantly down-regulated in the Cur group compared with the DC group (P<0.001, P<0.05, respectively) except of the protein content of PARP in the Cur group that was still obviously higher than that in the NC group (P<0.01).

Figure 2 The of the PKC and PARP protein levels and a diagram of curcumin attenuate oxidative stress. A, protein kinase C (PKC); B, poly ADP ribose polymerase (PARP); C, a diagram of curcumin attenuate oxidative stress. Data is presented as Means ± SE, indicates, * P<0.05, ** P<0.01 and *** P<0.001 means differences compared with NC group; # P<0.05, ## P<0.01, ### P<0.001 means differences compared with DC group. The abbreviations are as following: OS, oxidative stress; MDA, malondialdehyde; SOD, superoxide dismutase; GSH, reduced glutathione; GPx, glutathione peroxidase; SDH, sorbitol dehydrogenase.
Figure 2

The of the PKC and PARP protein levels and a diagram of curcumin attenuate oxidative stress. A, protein kinase C (PKC); B, poly ADP ribose polymerase (PARP); C, a diagram of curcumin attenuate oxidative stress. Data is presented as Means ± SE, indicates, * P<0.05, ** P<0.01 and *** P<0.001 means differences compared with NC group; # P<0.05, ## P<0.01, ### P<0.001 means differences compared with DC group. The abbreviations are as following: OS, oxidative stress; MDA, malondialdehyde; SOD, superoxide dismutase; GSH, reduced glutathione; GPx, glutathione peroxidase; SDH, sorbitol dehydrogenase.

4 Discussion

In this study, the major findings are described in the following section. Firstly, curcumin attenuated the NADH/ NAD+ redox imbalance in diabetic rats by inhibiting the polyol pathway and PARP. Secondly, curcumin treatment noticeably inhibited the PKC protein expression and increased the activities of glucose-polymerizing enzyme and glucose synthetase, resulting in a decreased level of oxidative stress. In our experiments, the blood glucose was markedly elevated in the DC group compared with the NC group, but the blood glucose level was attenuated significantly in the Cur group (Table 1). This results are consistent with the conclusions of previous research that showed that dietary supplements for curcumin could regulate glucose levels [22,35]. In this study, the results showed that curcumin treatment significantly increased the hepatic glycogen level compared with the DC group (Table 2), which is ascribed to the activities of glucose synthetase and increased glucose-polymerizing enzyme (Figure 1D and 1E). Previous studies have reported stress-sensitive kinases to be involved in insulin receptor substrate mediated insulin resistance in diabetes, including several isozymes of PKC-β and PKC-γ [36,37]. Activation of PKC isoforms are able to phosphorylate multiple targets associated with glycogen decomposition [2], along with increased hexosamine pathway flux [37]. In this study, curcumin treatment obviously down-regulated the PKC protein expression, suggesting that curcumin could increase the synthesis of glycogen regulator of blood glucose homeostasis.

A previous study demonstrated that oxidative stress could be prevented by the scavenging activity of antioxidant enzymes such as SOD and GSH [29,35,38,39,40]. GSH plays a critical role in regulating the intracellular redox system to defend cells from oxidative stress [41]. Recently, reports have shown that increased SOD and GSH activity could protect pancreatic β-cells from oxidative stress during diabetic conditions [6,42]. In the present study, plasma MDA was significantly increased in the DC group compared to those in the control group, indicating antioxidant defense system impairment. The activity of SOD was enhanced in the present study and may contribute to the protective effect of curcumin in diabetic rats by scavenging oxygen free radicals or enhancing the antioxidant capacity [6,42]. During diabetic condition, free radical production was increased due to chronic hyperglycemia which is involved in the β-cell dysfunction [6,7,43], and which could lead to β-cell death depending on the DNA alkylation, poly (ADP) ribose synthetase activation and redox imbalance [6,7,15,44]. The redox balance is dependent on activating two enzyme systems, including aldose reductase and sorbitol dehydrogenase [45]. Meanwhile, NADH/NAD+ redox balance was disrupted in the diabetic, resulting in their susceptibility to free radical damage [46]. Administration of curcumin to diabetic rats could attenuate these changes. Our study demonstrates that not only NADPH levels were increased but also G6PD activity and GSH levels were elevated in the curcumin-treated rat model. It seems that the increase in NADPH content in the Cur group (Fig. 1B) would increase GSH formation because NADPH is required for GSH formation from GSSG by glutathione reductase [47]. Our results showed that G6PD activity, NADPH and GSH levels in the Cur group were higher than in the DC group. These results are similar to the previous descriptions [48,49,50,51,52].

SDH is an important enzyme in the polyol pathway, which catalyzes the conversion of sorbitol to fructose in the presence of NAD. Previous results have shown that the activity of SDH was elevated in the diabetic, leading to increased availability of fructose and better substrate than glucose for glycosylation [1]. In the present research, the activity of SDH in the plasma and liver was decreased in the Cur group compared with the DC group. These changes can decrease availability of sorbitol to inhibit the cascade amplification effect of oxidative stress via negative feedback regulation of the SDH activity in diabetic rats. These results imply that curcumin may prevent diabetic pathological conditions by enhancing glycogen synthesis and reducing hyperglycemia in the diabetic rat model.

In conclusion, a diagram illustrating the proposed mechanism of action of curcumin is shown in Figure 2C. We propose that the effect of curcumin attenuates oxidative stress in STZ-induced diabetes by inhibiting polyol pathway, PARP expression and down-regulating expression of protein kinase C.

Acknowledgments

This work was supported by the Initial Funding of Jinshan College of Fujian Agriculture and Forestry University (Grant number Z150701), Scientific Research Foundation for Young and Middle-aged Teachers of Fujian Province (Grant number JAT160686) and Funding of Collaborative Innovation Center of Animal Health and Food Safety Application Technology in Fujian (Grant number 201703).

  1. Authors and Contributors: Substantial contributions to the conception or design of the work: ZL Xie; The acquisition data for the work: XQ Zeng, XQ Li, BB Wu, GZ Shen and QY Wu; Analysis data for the work: ZL Xie, XQ Zeng, XQ Li, BB Wu, GZ Shen and QY Wu; Interpretation of data for the work: ZL Xie; Drafting the work: ZL Xie and BB Wu; Revising it critically for important intellectual content: XQ Zeng, XQ Li, BB Wu, GZ Shen and QY Wu; Revised the manuscript: CB Wu. Final approval of the version to be published: ZL Xie, XQ Zeng, XQ Li, BB Wu, GZ Shen, QY Wu and CB Wu.

  2. Conflict of interest: Authors state no conflict of interest

References

[1] Brownlee M., Glycation products and the pathogenesis of diabetic complications, Diabetes care, 1992, 15(12), 1835-184310.2337/diacare.15.12.1835Search in Google Scholar PubMed

[2] Brownlee M., Biochemistry and molecular cell biology of diabetic complications, Nature, 2001, 414(6865), 813-82010.1038/414813aSearch in Google Scholar PubMed

[3] Gao Q., Shen W., Qin W., Zheng C., Zhang M., Zeng C., et al., Treatment of db/db diabetic mice with triptolide: a novel therapy for diabetic nephropathy, Nephrology, dialysis, transplantation: official publication of the European Dialysis and Transplant Association - European Renal Association, 2010, 25(11), 3539-4710.1093/ndt/gfq245Search in Google Scholar PubMed

[4] Kume S., Koya D., Uzu T., Maegawa H., Role of nutrient-sensing signals in the pathogenesis of diabetic nephropathy, BioMed research international, 2014, 31549410.1155/2014/315494Search in Google Scholar PubMed PubMed Central

[5] Luo X., Wu J., Jing S., Yan L.J., Hyperglycemic Stress and Carbon Stress in Diabetic Glucotoxicity, Aging and disease, 2016, 7(1), 90-11010.14336/AD.2015.0702Search in Google Scholar PubMed PubMed Central

[6] Gerber P.A., Rutter G.A., The Role of Oxidative Stress and Hypoxia in Pancreatic Beta-Cell Dysfunction in Diabetes Mellitus, Antioxidants & redox signaling, 201610.1089/ars.2016.6755Search in Google Scholar PubMed PubMed Central

[7] Kajimoto Y., Kaneto H., Role of oxidative stress in pancreatic beta-cell dysfunction, Annals of the New York Academy of Sciences, 2004, 1011, 168-17610.1196/annals.1293.017Search in Google Scholar

[8] Maritim A.C., Sanders R.A., Watkins J.B. 3rd, Diabetes, oxidative stress, and antioxidants: a review, Journal of biochemical and molecular toxicology, 2003, 17(1), 24-3810.1002/jbt.10058Search in Google Scholar PubMed

[9] Karunakaran U., Park K.G., A systematic review of oxidative stress and safety of antioxidants in diabetes: focus on islets and their defense, Diabetes & metabolism journal, 2013, 37(2), 106-11210.4093/dmj.2013.37.2.106Search in Google Scholar PubMed PubMed Central

[10] Jimenez-Flores L.M., Lopez-Briones S., Macias-Cervantes M.H., Ramirez-Emiliano J., Perez-Vazquez V., A PPARgamma, NF-kappaB and AMPK-dependent mechanism may be involved in the beneficial effects of curcumin in the diabetic db/db mice liver, Molecules 2014, 19(6), 8289-30210.3390/molecules19068289Search in Google Scholar PubMed PubMed Central

[11] Wang K., Molecular mechanisms of hepatic apoptosis regulated by nuclear factors, Cellular signalling, 2015, 27(4), 729-73810.1016/j.cellsig.2014.11.038Search in Google Scholar

[12] Tang W.H., Martin K.A., Hwa J., Aldose reductase, oxidative stress, and diabetic mellitus, Frontiers in pharmacology, 2012, 3, 8710.3389/fphar.2012.00087Search in Google Scholar

[13] Kador P.F., Kinoshita J.H., Role of aldose reductase in the development of diabetes-associated complications, The American journal of medicine, 1985, 79(5A), 8-1210.1016/0002-9343(85)90504-2Search in Google Scholar

[14] Yabe-Nishimura C., Aldose reductase in glucose toxicity: a potential target for the prevention of diabetic complications, Pharmacological reviews, 1998, 50(1), 21-33Search in Google Scholar

[15] Obrosova I.G., Drel V.R., Pacher P., Ilnytska O., Wang Z.Q., Stevens M.J., et al., Oxidative-nitrosative stress and poly(ADP-ribose) polymerase (PARP) activation in experimental diabetic neuropathy: the relation is revisited, Diabetes 2005, 54(12), 3435-344110.2337/diabetes.54.12.3435Search in Google Scholar PubMed PubMed Central

[16] Yan L.J., Pathogenesis of chronic hyperglycemia: from reductive stress to oxidative stress, Journal of diabetes research, 2014, 2014, 13791910.1155/2014/137919Search in Google Scholar PubMed PubMed Central

[17] Lieben L., Diabetic nephropathy: Lipid toxicity drives renal disease, Nature reviews Nephrology 2017, 13(4), 19410.1038/nrneph.2017.22Search in Google Scholar PubMed

[18] Flyvbjerg A., The role of the complement system in diabetic nephropathy, Nature reviews Nephrology, 201710.1038/nrneph.2017.31Search in Google Scholar PubMed

[19] Yamashita K., Takahiro K., Kamezaki F., Adachi T., Tasaki H., Decreased plasma extracellular superoxide dismutase level in patients with vasospastic angina, Atherosclerosis, 2007, 191(1), 147-15210.1016/j.atherosclerosis.2006.03.008Search in Google Scholar PubMed

[20] Li J.Y., Tao F., Wu X.X., Tan Y.Z., He L., Lu H., Polymorphic variations in manganese superoxide dismutase (MnSOD) and endothelial nitric oxide synthase (eNOS) genes contribute to the development of type 2 diabetes mellitus in the Chinese Han population, Genetics and molecular research, GMR, 2015, 14(4), 12993-300210.4238/2015.October.21.20Search in Google Scholar PubMed

[21] Chen H., Yu M., Li M., Zhao R., Zhu Q., Zhou W., et al., Polymorphic variations in manganese superoxide dismutase (MnSOD), glutathione peroxidase-1 (GPX1), and catalase (CAT) contribute to elevated plasma triglyceride levels in Chinese patients with type 2 diabetes or diabetic cardiovascular disease, Molecular and cellular biochemistry, 2012, 363(1-2), 85-9110.1007/s11010-011-1160-3Search in Google Scholar PubMed

[22] Hsu C.H., Cheng A.L., Clinical studies with curcumin, Advances in experimental medicine and biology, 2007, 595, 471-48010.1007/978-0-387-46401-5_21Search in Google Scholar PubMed

[23] Gupta S.C., Patchva S., Koh W., Aggarwal B.B., Discovery of curcumin, a component of golden spice, and its miraculous biological activities, Clinical and experimental pharmacology & physiology, 2012, 39(3), 283-29910.1111/j.1440-1681.2011.05648.xSearch in Google Scholar PubMed PubMed Central

[24] Shishodia S., Molecular mechanisms of curcumin action: gene expression, Biofactors, 2013, 39(1), 37-5510.1002/biof.1041Search in Google Scholar PubMed

[25] Sikora-Polaczek M., Bielak-Zmijewska A., Sikora E., Molecular and cellular mechanisms of curcumin action--beneficial effect on organism, Postepy biochemii, 2011, 57(1), 74-84Search in Google Scholar

[26] Joe B., Vijaykumar M., Lokesh B.R., Biological properties of curcumin-cellular and molecular mechanisms of action, Critical reviews in food science and nutrition, 2004, 44(2), 97-11110.1080/10408690490424702Search in Google Scholar PubMed

[27] Pescosolido N., Giannotti R., Plateroti A.M., Pascarella A., Nebbioso M., Curcumin: therapeutical potential in ophthalmology,, Planta medica, 2014, 80(4), 249-25410.1055/s-0033-1351074Search in Google Scholar PubMed

[28] Shehzad A., Ha T., Subhan F., Lee Y.S., New mechanisms and the anti-inflammatory role of curcumin in obesity and obesity-related metabolic diseases, European journal of nutrition, 2011, 50(3), 151-16110.1007/s00394-011-0188-1Search in Google Scholar PubMed

[29] Martinez-Morua A., Soto-Urquieta M.G., Franco-Robles E., Zúñiga-Trujillo I., Campos-Cervantes A., Pérez-Vázquez V., et al., Curcumin decreases oxidative stress in mitochondria isolated from liver and kidneys of high-fat diet-induced obese mice, Journal of Asian natural products research, 2013, 15(8), 905-91510.1080/10286020.2013.802687Search in Google Scholar PubMed

[30] Bhatia N.K., Srivastava A., Katyal N., Jain N., Khan M.A., Kundu B., et al., Curcumin binds to the pre-fibrillar aggregates of Cu/ Zn superoxide dismutase (SOD1) and alters its amyloidogenic pathway resulting in reduced cytotoxicity, Biochimica et biophysica acta, 2015, 1854(5), 426-43610.1016/j.bbapap.2015.01.014Search in Google Scholar PubMed

[31] Golalipour M.J., Ghafari S., Kouri V., Kestkar A.A., Proliferation of the b -Cells of Pancreas in Diabetic Rats Treated with Urtica Dioica, International Journal of Morphology, 2010, 28, 399-40410.4067/S0717-95022010000200011Search in Google Scholar

[32] Han D.N., Zhang D.H., Wang L.P., Zhang Y.S., Protective effect of beta-casomorphin-7 on cardiomyopathy of streptozotocin-induced diabetic rats via inhibition of hyperglycemia and oxidative stress, Peptides, 2013, 44, 120-12610.1016/j.peptides.2013.03.028Search in Google Scholar PubMed

[33] Yin H., Miao J., Zhang Y., Protective effect of beta-casomorphin-7 on type 1 diabetes rats induced with streptozotocin, Peptides, 2010, 31(9), 1725-172910.1016/j.peptides.2010.05.016Search in Google Scholar

[34] Yan L.J., Levine R.L., Sohal R.S., Oxidative damage during aging targets mitochondrial aconitase, Proceedings of the National Academy of Sciences of the United States of America, 1997, 94(21), 11168-1117210.1073/pnas.94.21.11168Search in Google Scholar

[35] Rahimi H.R., Mohammadpour A.H., Dastani M., Jaafari M.R., Abnous K., Ghayour Mobarhan M., et al., The effect of nano-curcumin on HbA1c, fasting blood glucose, and lipid profile in diabetic subjects: a randomized clinical trial, Avicenna journal of phytomedicine, 2016, 6(5), 567-577Search in Google Scholar

[36] Dey D., Mukherjee M., Basu D., Datta M., Roy S.S., Bandyopadhyay A., et al., Inhibition of insulin receptor gene expression and insulin signaling by fatty acid: interplay of PKC isoforms therein, Cellular physiology and biochemistry: international journal of experimental cellular physiology, biochemistry, and pharmacology, 2005, 16(4-6), 217-22810.1159/000089847Search in Google Scholar

[37] Giacco F., Brownlee M., Oxidative stress and diabetic complications, Circulation research, 2010, 107(9), 1058-107010.1161/CIRCRESAHA.110.223545Search in Google Scholar

[38] He X., de Seymour J.V., Sulek K., Qi H., Zhang H., Han T.L., et al., Maternal hair metabolome analysis identifies a potential marker of lipid peroxidation in gestational diabetes mellitus, Acta diabetologica, 2016, 53(1), 119-12210.1007/s00592-015-0737-9Search in Google Scholar

[39] Afanasiev S.A., Kondratieva D.S., Rebrova T.Y., Batalov R.E., Popov S.V., Coupling of the Functional Stability of Rat Myocardium and Activity of Lipid Peroxidation in Combined Development of Postinfarction Remodeling and Diabetes Mellitus, Journal of diabetes research, 2016, 2016, 254868910.1155/2016/2548689Search in Google Scholar

[40] Motterlini R., Foresti R., Bassi R., Green C.J., Curcumin, an antioxidant and anti-inflammatory agent, induces heme oxygenase-1 and protects endothelial cells against oxidative stress, Free radical biology & medicine, 2000, 28(8), 1303-131210.1016/S0891-5849(00)00294-XSearch in Google Scholar

[41] Grant C.M., MacIver F.H., Dawes I.W., Glutathione is an essential metabolite required for resistance to oxidative stress in the yeast Saccharomyces cerevisiae, Current genetics, 1996, 29(6), 511-51510.1007/BF02426954Search in Google Scholar PubMed

[42] Jin L., Xue H.Y., Jin L.J., Li S.Y., Xu Y.P., Antioxidant and pancreas-protective effect of aucubin on rats with streptozotocin-induced diabetes, European journal of pharmacology, 2008, 582(1-3), 162-16710.1016/j.ejphar.2007.12.011Search in Google Scholar PubMed

[43] Szkudelski T., The mechanism of alloxan and streptozotocin action in B cells of the rat pancreas, Physiological research, 2001, 50(6), 537-546Search in Google Scholar

[44] Bennett R.A., Pegg A.E., Alkylation of DNA in rat tissues following administration of streptozotocin, Cancer research, 1981, 41(7), 2786-2790Search in Google Scholar

[45] Ng T.F., Lee F.K., Song Z.T., Calcutt N.A., Lee A.Y., Chung S.S., et al., Effects of sorbitol dehydrogenase deficiency on nerve conduction in experimental diabetic mice, Diabetes, 1998, 47(6), 961-96610.2337/diabetes.47.6.961Search in Google Scholar PubMed

[46] Lenzen S., Drinkgern J., Tiedge M., Low antioxidant enzyme gene expression in pancreatic islets compared with various other mouse tissues, Free radical biology & medicine, 1996, 20(3), 463-46610.1016/0891-5849(96)02051-5Search in Google Scholar

[47] Luo X., Li R., Yan L.J., Roles of Pyruvate, NADH, and Mitochondrial Complex I in Redox Balance and Imbalance in beta Cell Function and Dysfunction, Journal of diabetes research, 2015, 2015, 51261810.1155/2015/512618Search in Google Scholar PubMed PubMed Central

[48] Gorin Y., Wauquier F., Upstream regulators and downstream effectors of NADPH oxidases as novel therapeutic targets for diabetic kidney disease, Molecules and cells, 2015, 38(4), 285-296Search in Google Scholar

[49] Sedeek M., Nasrallah R., Touyz R.M., Hebert R.L., NADPH oxidases, reactive oxygen species, and the kidney: friend and foe, Journal of the American Society of Nephrology: JASN, 2013, 24(10), 1512-151810.1681/ASN.2012111112Search in Google Scholar PubMed PubMed Central

[50] Gill P.S., Wilcox C.S., NADPH oxidases in the kidney, Antioxidants & redox signaling, 2006, 8(9-10), 1597-160710.1089/ars.2006.8.1597Search in Google Scholar PubMed

[51] Ulusu N.N., Sahilli M., Avci A., Canbolat O., Ozansoy G., Ari N., et al., Pentose phosphate pathway, glutathione-dependent enzymes and antioxidant defense during oxidative stress in diabetic rodent brain and peripheral organs: effects of stobadine and vitamin E, Neurochemical research, 2003, 28(6), 815-82310.1023/A:1023202805255Search in Google Scholar

[52] Gupte R.S., Floyd B.C., Kozicky M., George S., Ungvari Z.I., Neito V., et al., Synergistic activation of glucose-6-phosphate dehydrogenase and NAD(P)H oxidase by Src kinase elevates superoxide in type 2 diabetic, Zucker fa/fa, rat liver, Free radical biology & medicine, 2009, 47(3), 219-22810.1016/j.freeradbiomed.2009.01.028Search in Google Scholar PubMed PubMed Central

Received: 2017-7-9
Accepted: 2017-10-30
Published Online: 2017-12-29

© 2017 Zhenglu Xie et al.

This work is licensed under the Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 License.

Downloaded on 25.5.2024 from https://www.degruyter.com/document/doi/10.1515/biol-2017-0053/html
Scroll to top button