Protective effects of Lagerstroemia speciosa on 3-morpholinosydnonimine (SIN-1)-induced oxidative stress in HIT-T15 pancreatic β cells

  • Authors:
    • Jia-Le Song
    • Xin Zhao
    • Qiang Wang
    • Ting Zhang
  • View Affiliations

  • Published online on: March 26, 2013     https://doi.org/10.3892/mmr.2013.1396
  • Pages: 1607-1612
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Reactive oxygen species (ROS)-induced pancreatic β cell death affects insulin secretion and is important in the pathogenesis of diabetes. Lagerstroemia speciosa, a traditional folk medicine, has been used for t he prevention and treatment of diabetes. However, whether Lagerstroemia speciosa has a cytoprotective effect on pancreatic β cells remains to be elucidated. The present study aimed to investigate the cytoprotective effects of hot water extracts from Lagerstroemia speciosa leaves (LWE) on 3-morpholinosydnonimine (SIN-1)-induced oxidative damage in Syrian hamster pancreatic insulinoma HIT-T15 cells. The HIT-T15 cells were first treated with SIN-1 (50 µM) for 24 h and then co-incubated with LWE for 48 h. SIN-1 significantly decreased HIT-T15 cell viability (P<0.05); however, LWE did not exert a significant cytotoxic effect and increased the viability of HIT-T15 cells in a dose‑dependent manner. To further investigate the protective effects of LWE on SIN-1‑induced oxidative stress in HIT-T15 cells, the cellular levels of ROS, lipid peroxidation and endogenous antioxidant enzymes, including superoxide dismutase (SOD), catalase (CAT) and glutathione peroxidase (GSH-px), were determined. LWE decreased the intracellular levels of ROS and lipid peroxidation, and increased the activities of antioxidant enzymes. These results suggest that LWE has a cytoprotective effect against SIN-1‑induced oxidative stress in HIT-T15 cells through the inhibition of lipid peroxidation, a decrease in ROS levels and an increase in antioxidant enzyme activity. In addition, LWE increased insulin secretion in SIN-1-treated HIT-T15 cells. Our results suggested that LWE were effective in the treatment of diabetes. Further studies are required to study the anti-diabetic molecular mechanism in a cell model.

Introduction

Diabetes mellitus has become an increasingly important public health problem worldwide. However, the etiology of diabetes mellitus has not yet been fully elucidated. Reactive oxygen species (ROS)-induced oxidative stress is known to be important in the pathogenic process of diabetes mellitus. ROS that are particularly responsible for oxidative stress include superoxide (O2), hydroxyl radical (OH), singlet oxygen (1O2), hydrogen peroxide (H2O2), nitric oxide (NO) and peroxynitrite (ONOO) (1). Oxidative stress may induce the dysfunction of pancreatic β cells, decreased insulin secretion (2) and the development of diabetic complications, including retinopathy, nephropathy, neuropathy and vascular damage (3,4). Generally, various antioxidative compounds exist in mammalian cells, including low-molecular mass antioxidants such as glutathione (GSH), uric acid, vitamin C, vitamin E and various endogenous antioxidant enzymes against oxidative stress. It is widely accepted that superoxide dismutase (SOD), catalase (CAT) and glutathione peroxidase (GSH-px) are three important endogenous antioxidant enzymes for the protection of living organs against ROS-induced oxidative stress. Among these antioxidant enzymes, SOD catalyzes the dismutation of O2 into H2O2, which may be transformed into H2O and O2 by CAT. GSH-px is crucial for removing lipid hydroperoxides and reducing free H2O2 to water (1).

A number of drugs used in clinical diabetes mellitus treatment have been associated with side-effects, including gastrointestinal disturbances, edema, myocardial infarction and risk of cardiovascular disease (58). To date, >400 traditional plant treatments for diabetes mellitus have been identified (9). The anti-diabetic components of these natural plants may constitute ancillary medication for diabetes treatment.

Lagerstroemia speciosa (Lythraceae), also named banaba, is a tropical plant that grows in several parts of southeast Asia, including southern China, Vietnam, Malaysia and the Philippines. Lagerstroemia speciosa has been used as a traditional folk medicine for the treatment of diabetes and kidney-related diseases in the Philippines for ~1,000 years (10,11). A number of studies have reported that Lagerstroemia speciosa has antioxidant (12,13), anti-inflammatory (13), anticancer (14), anti-obesity (15) and antidiabetic (12,16) activities. Tannic and triterpene acids are the main components of Lagerstroemia speciosa leaf extracts and have been shown to downregulate blood glucose and possess apparent antidiabetic properties in vivo and in vitro(1619).

The present study aimed to investigate the potential cytoprotective effects of hot water extracts from Lagerstroemia speciosa leaves (LWE) on 3-morpholinosydnonimine (SIN-1)-induced oxidative stress in HIT-T15 cells and to elucidate the underlying mechanisms involved in this process.

Materials and methods

Plant extract preparation

Fresh Lagerstroemia speciosa leaves were purchased from a local market in Chongqing, China. LWE was prepared by boiling 160 g air-dried Lagerstroemia speciosa leaves in 1 l distilled water for 2 h, followed by ultracentrifugation at 30,000 × g for 30 min, filtration with a 0.4-μm filter, concentration by heat evaporation and freeze-drying. LWE was redissolved in dimethyl sulfoxide (DMSO) at a concentration of 50 mg/ml and stored at 4°C until future use.

Cell culture

Syrian hamster insulin-secreting HIT-T15 cells were obtained from the American Type Culture Collection (ATCC, Manassas, VA, USA). The cells were routinely maintained in RPMI-1640 medium supplemented with 10% (v/v) fetal bovine serum (FBS) and 1% penicillin-streptomycin in a humidified CO2 incubator (model 3154; Forma Scientific, Inc., Marietta, OH, USA) with 5% CO2 at 37°C.

Cell viability assay

Cell viability was assessed using the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide (MTT) method. The cells were seeded in 96-well plates at a density of 5×103 cells/well. Following 24-h incubation, the cells were primarily treated with SIN-1 (50 μM) for 24 h and then incubated with LWE (1–100 μg/ml) for 48 h. Next, 100 μl MTT reagent (final concentration, 0.5 mg/ml) was added to each well and the cells were incubated in a humidified incubator at 37°C to allow MTT to be metabolized. After 4 h, 100 μl DMSO was added to each well to dissolve formazan deposits. The absorbance of the samples was measured at a wavelength of 540 nm using a microplate reader (model 680; Bio-Rad, Hercules, CA, USA).

Determination of intracellular ROS

Intracellular ROS levels were measured using the fluorescent probe dihydrodichlorofluorescein (H2DCFDA). Following treatment, HIT-T15 cells were washed with calcium- and magnesium-free phosphate-buffered saline (PBS) and incubated in H2DCFDA (20 μM) containing serum- and phenol red-free Dulbecco’s modified Eagle’s medium (DMEM) for 30 min. Following incubation, the medium was removed and cells were washed with PBS twice. Fluorescence was measured using a FLUOstar OPTIMA fluorescence plate reader (BMG Labtech, Ortenberg, Germany); excitation was read at 485 nm and emission at 535 nm. Relative ROS production (calculated as a percentage of the control) was expressed as the ratio of fluorescence in the treated samples over the response in the appropriate controls: (fluorescencetreatment/fluorescencecontrol) ×100.

Lipid peroxidation levels

Lipid peroxidation was evaluated using a thiobarbituric acid reactive substance (TBARS) assay (20). Briefly, the cultured cells were washed with cooled PBS, scraped into trichloroacetic acid (TCA; 2.8%, w/v) and sonicated; total protein was determined using a bicinchoninic acid (BCA) assay. The suspension was mixed with 1 ml TBA (0.67%, w/v) and 1 ml TCA (25%, w/v), heated (30 min at 95°C) and centrifuged (22,000 × g; 10 min at 4°C). TBA reacted with the oxidative degradation products of lipids, yielding red complexes that absorbed at 535 nm. The level of TBARS was determined using a UV-2401PC spectrophotometer (Shimadzu, Kyoto, Japan).

Antioxidant enzyme activity

HIT-T15 cells grown in a 10-cm cell culture dish were co-incubated with SIN-1 (50 μM) for 24 h and then treated with LWE (2.5–50 μg/ml) for 48 h for further assessment. The cells were washed with PBS, removed by scraping and centrifuged, and the resulting cell pellet was stored at −80°C. Cell pellets were thawed, resuspended in 300 ml cold lysis buffer (PBS and 1 mM EDTA), homogenized and centrifuged (22,000 × g; 10 min at 4°C). The resulting supernatants were used for activity measurements. CAT activity (U/mg protein) was assessed according to the method described by Nelson and Kiesow (21), in which the disappearance of the substrate H2O2 was measured spectophotometrically at 240 nm. SOD activity (U/mg protein) was assayed using a modified auto-oxidation of pyrogallol method (22). One unit of SOD activity was defined as the amount of enzyme that inhibited the auto-oxidation rate of pyrogallol by 50%. GSH-px activity (U/mg protein) was assayed according to the method described by Hafemen et al(23). Protein contents were determined using the Bio-Rad protein assay kit according to the manufacturer’s instructions.

Insulin secretion assay

Insulin secretion was measured using a radioimmunoassay (RIA) method. The cells were seeded at a density of 5×105 cells/well in a 96-well plate and primarily treated with SIN-1 (50 μM) for 24 h, followed by LWE (2.5–50 μg/ml) for 48 h. To determine the level of insulin secreted, aliquots of samples (10 μl/well) were collected from the experimental medium at the indicated time points (48 h) and subjected to an insulin antiserum immunoassay within 5 min, according to the manufacturer’s instructions (Linco Research, Inc., St. Charles, MO, USA). The absorbance was read at 450 and 590 nm using a model 680 microplate reader (Bio-Rad) and results were recorded.

Statistical analysis

Data are presented as the mean ± SD. The differences between the mean values of individual groups were assessed using one-way ANOVA and Duncan’s multiple range tests. P<0.05 was considered to indicate a statistically significant difference. The SAS v9.1 statistical software package (SAS Institute, Inc., Cary, NC, USA) was used for analysis.

Results

Effects of LWE on SIN-1-induced oxidative damage in HIT-T15 cells

To investigate LWE-induced cytotoxicity, HIT-T15 cells were first treated with various concentrations of LWE (1–100 μg/ml) for 48 h and the cell viability was determined using an MTT assay. Treatment with LWE at doses of 1–50 μg/ml at 37°C for 48 h did not cause significant cytotoxicity and cell viability was >80%. A dose of 100 μg/ml LWE induced cell damage (cell viability, 76%; Fig. 1). Based on these results, concentrations of 2.5–50 μg/ml LWE were used for further assessment. As shown in Fig. 2, SIN-1 (50 μM) significantly induced cell death in HIT-T15 cells. However, following treatment with various concentrations of LWE, the cell viability increased in a dose-dependent manner.

Effects of LWE against SIN-1-induced intracellular ROS levels in HIT-T15 cells

To investigate the protective effects of LWE in SIN-1-treated HIT-T15 cells, the intracellular ROS levels were determined using the fluorescent probe H2DCFDA. As shown in Fig. 3, SIN-1 significantly increased ROS levels compared with those in the control cells. In the presence of SIN-1, LWE at doses of 2.5–50 μg/ml significantly reduced ROS generation in a dose-dependent manner. The intracellular ROS levels were 203.2±14.6, 194.3±9.9, 188.3±9.8, 178.3±7.5 and 175.7±10.8% when the cells were treated with 2.5, 5, 10, 25 and 50 μg/ml LWE, respectively. Treatment with the same concentrations of LWE alone did not significantly increase the intracellular ROS levels (date not shown). These results suggest that LWE is a free radical scavenger.

Effects of LWE on lipid peroxidation in SIN-1-treated HIT-T15 cells

Free radical- and ROS-induced oxidative damage has been strongly associated with the lipid peroxidation of cell membranes and increased levels of malondialdehyde (MDA), which is a biomarker of cell membrane lipid peroxidation. As shown in Fig. 4, SIN-1 significantly increased the level of MDA (1.23±0.10 nmol/mg protein) compared with that in the control cells (0.33±0.05 nmol/mg protein). LWE at doses of 2.5–50 μg/ml significantly reduced MDA levels in a dose-dependent manner. The MDA levels were 0.91±0.15, 0.87±0.05, 0.79±0.05, 0.69±0.01 and 0.58±0.02 nmol/mg protein when the cells were treated with 2.5, 5, 10, 25 and 50 μg/ml LWE, respectively.

Effects of LWE on the activity of antioxidant enzymes in SIN-1-treated HIT-T15 cells

Figs. 57 demonstrate the intracellular antioxidant enzyme activities with LWE in SIN-1-treated HIT-T15 cells. The activity of SOD was decreased with SIN-1 treatment (4.96±0.46 U/mg protein) compared with that in the control cells; however, this recovered to 5.39±0.83, 6.13±0.78, 7.19±0.26, 7.37±0.99 and 8.04±0.73 U/mg protein when the cells were treated with 2.5, 5, 10, 25 and 50 μg/ml LWE, respectively. Following treatment with SIN-1, cellular CAT was decreased (0.86±0.10 U/mg protein) compared with that in the control cells (1.83±0.21 U/mg protein). However, CAT activity was significantly increased (P<0.05) following treatment with LWE (Fig. 6). Additionally, LWE reduced the SIN-1-induced decrease in GSH-px in HIT-T15 cells. The GSH-px activity was identified to be significantly increased from 1.93±0.17 to 2.63±0.17 U/mg protein when the cells were treated with LWE (Fig. 7).

Effects of LWE on insulin secretion in SIN-1-treated HIT-T15 cells

As shown in Fig. 8, SIN-1 significantly decreased insulin levels (2095.4±105.0 pg/ml) compared with those in the control cells (10236.7±98.9 pg/ml). Following treatment with 2.5, 5, 10, 25 and 50 μg/ml LWE, the insulin levels were 2433.7±34.5, 2824.2±150.3, 3565.4±223.3, 4730.9±140.3 and 5069.2±131.5 pg/ml, respectively. These results suggest that LWE treatment is effective in increasing pancreatic β cell survival and maintaining normal biological functions in ROS-induced diabetes.

Discussion

ROS-induced oxidative damage in pancreatic β cells is considered to be important in the pathological process of diabetes. A number of studies have shown that reducing ROS levels and treatment with antioxidants (including NAC, vitamin C and vitamin E) improved β cell structure and function in vitro(24,25). However, whether LWE protects pancreatic β cells against SIN-1-induced oxidative damage has not yet been elucidated. In the present study, we demonstrated that LWE protected HIT-T15 cells against ROS-induced cell damage. The cytoprotective effects of LWE were mainly mediated by increased intracellular antioxidant enzyme activity.

The results of this study clearly showed that LWE prevented SIN-1-induced cell death, as assessed using the MTT assay. Additionally, LWE alone was not significantly cytotoxic to cells at the concentrations used. Treatment with LWE was shown to have a significant protective effect, which may be attributed to the free radical scavenging activity of the extract.

To evaluate the role of free radicals in the protective activity of LWE, the effect of LWE on SIN-1-induced ROS generation was analyzed using the H2DCFDA assay. SIN-1 treatment alone significantly increased intracellular ROS generation. Following treatment with LWE, ROS generation was found to decline in a dose-dependent manner. This decrease in the SIN-1-induced ROS generation may account for the decline in the observed cytoprotective effect of LWE.

Lipid peroxidation is the most extensively investigated process induced by free radicals. ROS participate in the toxic actions that lead to apoptosis in insulin-producing cells (26). In the present study, increased lipid peroxidation levels were observed in SIN-1-treated HIT-T15 cells. However, treatment with LWE resulted in a decrease in lipid peroxidation, indicating that oxidative stress-related damage was reduced in LWE-treated cells. The ability of LWE to reduce lipid peroxidation may be due to its function as a preventive antioxidant to scavenge initiating radicals.

The overproduction and consequently increased levels of free radicals may be scavenged by endogenous antioxidant enzymes, including SOD and GSH-px. In cells, SOD catalyzes the conversion of O2 to H2O2, and H2O2 is further reduced to H2O by the activity of CAT or GSH-px. Pancreatic β cells have been reported to contain low levels of endogenous antioxidant enzymes, particularly GSH-px and CAT (27). In the present study, SIN-1-treated HIT-T15 cells were shown to have decreased GSH-px and CAT activities, which may be due to the increased oxidative damage induced by SIN-1. However, LWE treatment caused an increase in the activity of these antioxidant enzymes in HIT-T15 cells, indicating that LWE reduced SIN-1-induced oxidative stress. A number of studies have reported that the overexpression of CuZnSOD had a protective effect in NO-induced human islets, INS-1 insulin-secreting cells (28) and alloxan- and streptozotocin-induced diabetes (29,30). CAT has also exhibited a protective effect against H2O2 and streptozotocin-induced oxidative stress in vivo(31). Additionally, combinatorial overexpression of CAT and GSH-px has been shown to have a protective effect against ROS-induced oxidative stress through improving the activity of CuZnSOD or MnSOD (3235).

In conclusion, the present study showed that LWE had protective activity against SIN-1-induced cell death in Syrian hamster HIT-T15 insulin-secreting cells. LWE effectively scavenged the products of SIN-1-induced intracellular ROS generation and reduced pancreatic β cell death through increasing the activity of intracellular antioxidant enzymes, including SOD, CAT and GSH-Px. LWE also promoted insulin secretion in SIN-1-treated HIT-T15 cells.

Acknowledgements

This study was supported by the Natural Science Foundation Project of CQ CSTC (No. CSTC2012jjA80002) and the Science and Technology Research Project of Chongqing Municipal Education Commission (No. KJ121504)

References

1 

Halliwell B: Reactive species and antioxidants. Redox biology is a fundamental theme of aerobic life. Plant Physiol. 141:312–22. 2006. View Article : Google Scholar : PubMed/NCBI

2 

Evans JL, Goldfine ID, Maddux BA and Grodsky GM: Are oxidative stress-activated signaling pathways mediators of insulin resistance and beta-cell dysfunction? Diabetes. 52:1–8. 2003. View Article : Google Scholar : PubMed/NCBI

3 

Rahimi R, Nikfar S, Larijani B and Abdollahi M: A review on the role of antioxidants in the management of diabetes and its complications. Biomed Pharmacother. 59:365–373. 2005. View Article : Google Scholar : PubMed/NCBI

4 

Robertson RP and Harmon JS: Diabetes, glucose toxicity, and oxidative stress: a case of double jeopardy for the pancreatic islet beta cell. Free Radic Biol Med. 41:177–184. 2006. View Article : Google Scholar : PubMed/NCBI

5 

Bell DS: Do sulfonylurea drugs increase the risk of cardiac events? CMAJ. 174:185–186. 2006. View Article : Google Scholar : PubMed/NCBI

6 

Home PD, Pocock SJ, Beck-Nielsen H, Gomis R, Hanefeld M, Jones NP, Komajda M and McMurray JJ; RECORD Study Group. Rosiglitazone evaluated for cardiovascular outcomes - an interim analysis. N Engl J Med. 357:28–38. 2007. View Article : Google Scholar : PubMed/NCBI

7 

Psaty BM and Furberg CD: The record on rosiglitazone and the risk of myocardial infarction. N Engl J Med. 357:67–69. 2007. View Article : Google Scholar : PubMed/NCBI

8 

Nathan DM: Rosiglitazone and cardiotoxicity - weighing the evidence. N Engl J Med. 357:64–66. 2007. View Article : Google Scholar : PubMed/NCBI

9 

Bailey CJ and Day C: Traditional plant medicines as treatments for diabetes. Diabetes Care. 12:553–564. 1989. View Article : Google Scholar : PubMed/NCBI

10 

Garcia F: Distribution and deterioration of insulin-like principle in Lagerstroemia speciosa (banaba). Acta Med Philippina. 3:99–104. 1941.

11 

Klein G, Kim J, Himmeldirk K, Cao Y and Chen X: Antidiabetes and anti-obesity activity of Lagerstroemia speciosa. Evid Based Complement Alternat Med. 4:401–407. 2007. View Article : Google Scholar

12 

Mishra Y, Khan MSY, Zafar R and Agarwal SS: Hypoglycemic activity of leaves of Lagerstroemia speciosa (L) Pers. Indian J Pharmacol. 22:174–176. 1990.

13 

Priya TT, Sabu MC and Jolly CI: Free radical scavenging and anti-inflammatory properties of Lagerstroemia speciosa (L). Inflammopharmacology. 16:182–187. 2008. View Article : Google Scholar : PubMed/NCBI

14 

Khan MT, Lampronti I, Martello D, Bianchi N, Jabbar S, Choudhuri MS, Datta BK and Gambari R: Identification of pyrogallol as an antiproliferative compound present in extracts from the medicinal plant Emblica officinalis: effects on in vitro cell growth of human tumor cell lines. Int J Oncol. 21:187–192. 2002.PubMed/NCBI

15 

Suzuki Y, Unno T, Ushitani M, Hayashi K and Kakuda T: Antiobesity activity of extracts from Lagerstroemia speciosa L. leaves on female KK-Ay mice. J Nutr Sci Vitaminol (Tokyo). 45:791–795. 1999. View Article : Google Scholar

16 

Kakuda T, Sakane I, Takihara T, Ozaki Y, Takeuchi H and Kuroyanagi M: Hypoglycemic effect of extracts from Lagerstroemia speciosa L. leaves in genetically diabetic KK-AY mice. Biosci Biotechnol Biochem. 60:204–208. 1996.

17 

Liu F, Kim J, Li Y, Liu X, Li J and Chen X: An extract of Lagerstroemia speciosa L. has insulin-like glucose uptake-stimulatory and adipocyte differentiation-inhibitory activities in 3T3-L1 cells. J Nutr. 131:2242–2247. 2001.PubMed/NCBI

18 

Liu X, Kim JK, Li Y, Li J, Liu F and Chen X: Tannic acid stimulates glucose transport and inhibits adipocyte differentiation in 3T3-L1 cells. J Nutr. 135:165–171. 2005.PubMed/NCBI

19 

Liu J, Sun H, Duan W, Mu D and Zhang L: Maslinic acid reduces blood glucose in KK-Ay mice. Biol Pharm Bull. 30:2075–2078. 2007. View Article : Google Scholar : PubMed/NCBI

20 

Fraga CG, Leibovitz BE and Tappel AL: Lipid peroxidation measured as thiobarbituric acid-reactive substances in tissue slices: characterization and comparison with homogenates and microsomes. Free Radic Biol Med. 4:155–161. 1988. View Article : Google Scholar

21 

Nelson DP and Kiesow LA: Enthalpy of decomposition of hydrogen peroxide by catalase at 25 degrees C (with molar extinction coefficients of H2O2 solutions in the UV). Anal Biochem. 49:474–478. 1972. View Article : Google Scholar : PubMed/NCBI

22 

Marklund S and Marklund G: Involvement of the superoxide anion radical in the autoxidation of pyrogallol and a convenient assay for superoxide dismutase. Eur J Biochem. 47:469–474. 1974. View Article : Google Scholar : PubMed/NCBI

23 

Hafeman DG, Sunde RA and Hoekstra WG: Effect of dietary selenium on erythrocyte and liver glutathione peroxidase in the rat. J Nutr. 104:580–587. 1974.PubMed/NCBI

24 

Robertson RP, Harmon J, Tran PO, Tanaka Y and Takahashi H: Glucose toxicity in beta-cells: type 2 diabetes, good radicals gone bad, and the glutathione connection. Diabetes. 52:581–587. 2003. View Article : Google Scholar : PubMed/NCBI

25 

Cheng Q, Law PK, de Gasparo M and Leung PS: Combination of the dipeptidyl peptidase IV inhibitor LAF237 [(S)-1-[(3-hydroxy-1-adamantyl)ammo]acetyl-2-cyanopyrrolidine] with the angiotensin II type 1 receptor antagonist valsartan [N-(1-oxopentyl)-N-[[2′-(1H-tetrazol-5-yl)-[1,1′-biphenyl]-4-yl]methyl]-L-valine] enhances pancreatic islet morphology and function in a mouse model of type 2 diabetes. J Pharmacol Exp Ther. 327:683–691. 2008.

26 

Lenzen S: Oxidative stress: the vulnerable beta-cell. Biochem Soc Trans. 36:343–347. 2008. View Article : Google Scholar : PubMed/NCBI

27 

Zhang H, Ollinger K and Brunk U: Insulinoma cells in culture show pronounced sensitivity to alloxan-induced oxidative stress. Diabetologia. 38:635–641. 1995. View Article : Google Scholar : PubMed/NCBI

28 

Moriscot C, Pattou F, Kerr-Conte J, Richard MJ, Lemarchand P and Benhamou PY: Contribution of adenoviral-mediated superoxide dismutase gene transfer to the reduction in nitric oxide-induced cytotoxicity on human islets and INS-1 insulin-secreting cells. Diabetologia. 43:625–631. 2000. View Article : Google Scholar

29 

Kubisch HM, Wang J, Bray TM and Phillips JP: Targeted overexpression of Cu/Zn superoxide dismutase protects pancreatic beta-cells against oxidative stress. Diabetes. 46:1563–1566. 1997. View Article : Google Scholar : PubMed/NCBI

30 

Kubisch HM, Wang J, Luche R, Carlson E, Bray TM, Epstein CJ and Phillips JP: Transgenic copper/zinc superoxide dismutase modulates susceptibility to type I diabetes. Proc Natl Acad Sci USA. 91:9956–9959. 1994. View Article : Google Scholar : PubMed/NCBI

31 

Xu B, Moritz JT and Epstein PN: Overexpression of catalase provides partial protection to transgenic mouse beta cells. Free Radic Biol Med. 27:830–837. 1999. View Article : Google Scholar : PubMed/NCBI

32 

Amstad P, Moret R and Cerutti P: Glutathione peroxidase compensates for the hypersensitivity of Cu,Zn-superoxide dismutase overproducers to oxidant stress. J Biol Chem. 269:1606–1609. 1994.PubMed/NCBI

33 

Lortz S and Tiedge M: Sequential inactivation of reactive oxygen species by combined overexpression of SOD isoforms and catalase in insulin-producing cells. Free Radic Biol Med. 34:683–688. 2003. View Article : Google Scholar : PubMed/NCBI

34 

Lepore DA, Shinkel TA, Fisicaro N, Mysore TB, Johnson LE, d’Apice AJ and Cowan PJ: Enhanced expression of glutathione peroxidase protects islet beta cells from hypoxia-reoxygenation. Xenotransplantation. 11:53–59. 2004. View Article : Google Scholar : PubMed/NCBI

35 

Mysore TB, Shinkel TA, Collins J, Salvaris EJ, Fisicaro N, Murray-Segal LJ, Johnson LE, Lepore DA, Walters SN, Stokes R, Chandra AP, O’Connell PJ, d’Apice AJ and Cowan PJ: Overexpression of glutathione peroxidase with two isoforms of superoxide dismutase protects mouse islets from oxidative injury and improves islet graft function. Diabetes. 54:2109–2116. 2005. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

May 2013
Volume 7 Issue 5

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Song J, Zhao X, Wang Q and Zhang T: Protective effects of Lagerstroemia speciosa on 3-morpholinosydnonimine (SIN-1)-induced oxidative stress in HIT-T15 pancreatic β cells. Mol Med Rep 7: 1607-1612, 2013
APA
Song, J., Zhao, X., Wang, Q., & Zhang, T. (2013). Protective effects of Lagerstroemia speciosa on 3-morpholinosydnonimine (SIN-1)-induced oxidative stress in HIT-T15 pancreatic β cells. Molecular Medicine Reports, 7, 1607-1612. https://doi.org/10.3892/mmr.2013.1396
MLA
Song, J., Zhao, X., Wang, Q., Zhang, T."Protective effects of Lagerstroemia speciosa on 3-morpholinosydnonimine (SIN-1)-induced oxidative stress in HIT-T15 pancreatic β cells". Molecular Medicine Reports 7.5 (2013): 1607-1612.
Chicago
Song, J., Zhao, X., Wang, Q., Zhang, T."Protective effects of Lagerstroemia speciosa on 3-morpholinosydnonimine (SIN-1)-induced oxidative stress in HIT-T15 pancreatic β cells". Molecular Medicine Reports 7, no. 5 (2013): 1607-1612. https://doi.org/10.3892/mmr.2013.1396