Exp Clin Endocrinol Diabetes 2017; 125(09): 583-591
DOI: 10.1055/s-0035-1569332
Review
© Georg Thieme Verlag KG Stuttgart · New York

Effects of Low Intensity Exercise Against Apoptosis and Oxidative Stress in Streptozotocin-induced Diabetic Rat Heart

M. Kanter
1   Department of Histology and Embryology, Faculty of Medicine, Istanbul Medeniyet University, Istanbul, Turkey
,
F. Aksu
2   Department of Cardiology, Istanbul Medeniyet University, Goztepe Education and Research Hospital, Istanbul/Turkey
,
M. Takir
3   Clinics of Internal Medicine and Endocrinology, Istanbul Medeniyet University, Goztepe Education and Research Hospital, Istanbul/Turkey
,
O. Kostek
4   Clinics of Internal Medicine, Istanbul Medeniyet University, Goztepe Education and Research Hospital, Istanbul/Turkey
,
B. Kanter
5   Faculty of Medicine, Medical University, Sofia, Bulgaria
,
A. Oymagil
6   Department of Pediatry, Istanbul Medeniyet University, Goztepe Education and Research Hospital, Istanbul/Turkey
› Author Affiliations
Further Information

Publication History

received     06 October 2015
first decision  09 November 2015

accepted     03 December 2015

Publication Date:
29 January 2016 (online)

Abstract

Background The aim of this study was to investigate the effects of low intensity exercise on heart of streptozotocin (STZ)-induced diabetic rats.

Materials and Methods The rats were randomly divided into 3 experimental groups: A (control), B (diabetic untreated), and C (diabetic treated with low intensity exercise); each group contains 8 animals. B and C groups received STZ. Diabetes was induced in 2 groups by a single intraperitoneal (i.p) injection of STZ (40 mg/kg, freshly dissolved in 0,1 M citrate buffer, pH 4.2). 2 days after STZ treatment, diabetes in 2 experimental groups was confirmed by measuring blood glucose levels. Rats with blood glucose levels of 250 mg/dl or higher were considered to be diabetic. Animals in the exercise group were made to run the treadmill once a day for 4 consecutive weeks. Exercise started 3 days prior to STZ administration.

Results After induction of diabetes, histological abnormalities were observed, including myofibrillar loss, vacuolization of cytoplasm and irregularity of myofibrils. These alterations were attenuated by low intensity exercise. Our data indicates a significant reduction of oxidative stress and apoptosis in cardiomyocytes after exercise. Treatment of diabetic animals with low intensity exercise, decreased the elevated tissue malondialdehyde (MDA) levels and increased the reduced activities of the enzymatic antioxidants superoxide dismutase (SOD), glutathione peroxidase (GSH-Px), and catalase (CAT) in cardiac tissue.

Conclusion These findings suggest that low intensity exercise has a therapeutic protective effect in diabetes by decreasing oxidative stress and apoptosis, and by preservation of myocardial integrity.

 
  • References

  • 1 Weiss RB. Streptozotocin: a review of it pharmacology, efficacy and toxicity. Cancer Treat Rep 1982; 66: 427-438
  • 2 Uchigata Y, Yamamoto H, Kawamura A. et al. Protection by superoxide dismutase, catalase and poly (ADPribose) sunthetase inhibitors against alloxan-and streptozotocin induced islet DNA strand breaks and against the inhibition of proinsulin synthesis. J Biol Chem 1982; 257: 6084-6088
  • 3 Merzouk H, Madani S, Chabane D. et al. Time-course of changes in serum glucose, insulin, lipids and tissue lipase activities in macrosomic offspring of rats with streptozotocin-induced diabetes. Clin Sci 2000; 98: 21-30
  • 4 Martínez-Ramonde T, Alonso N, Cordido F. et al. Importance of exercise in the control of metabolic and inflammatory parameters at the moment of onset in type 1 diabetic subjects. Exp Clin Endocrinol Diabetes 2014; 122: 334-340
  • 5 Li X, Jiang YH, Jiang P. et al. Analysis of Heart Rate Variability and Cardiac Autonomic Nerve Remodeling in Streptozotocin-induced Diabetic Rats. Exp Clin Endocrinol Diabetes 2015; 123: 272-281
  • 6 Chen YT, Zheng RL, Jia ZJ. et al. Flavonoids as superoxide scavengers and antioxidants. Free Radic Biol Med 1990; 9: 19-20
  • 7 Kroncke KD, Fehsel K, Sommer A. et al. Nitric oxide generation during cellular metabolization of the diabetogenic N-methyl-nitroso-urea: streptozotocin contributes to islet cell DNA damage. Biol Chem Hoppe-Seyler 1995; 376: 179-185
  • 8 Tiedge M, Lortz S, Drinkgern J. et al. Relation between antioxidant enzyme gene expression and antioxidative defense status of insulinproducing cells. Diabetes 1997; 46: 1733-1742
  • 9 Matkovics B, Kotorman M, Varga IS. et al. Oxidative stress in experimental diabetes induced by streptozotocin. Acta Physiol Hung 1998; 85: 29-38
  • 10 Go AS, Mozaffarian D, Roger VL. et al. Heart disease and stroke statistics – 2014 update: a report from the American Heart Association. Circulation 2014; 129: 399-410
  • 11 Rubler S, Dlugash J, Yuceoglu YZ. et al. New type of cardiomyopathy associated with diabetic glomerulosclerosis. Am J Cardiol 1972; 30: 595-602
  • 12 Miki T, Yuda S, Kouzu H. et al. Diabetic cardiomyopathy: pathophysiology and clinical features. Heart Fail Rev 2013; 18: 149-166
  • 13 Ward 1 ML, Crossman DJ. Mechanisms underlying the impaired contractility of diabetic cardiomyopathy. World J Cardiol 2014; 26 6: 577-584
  • 14 Zarain-Herzberg A, García-Rivas G, Estrada-Avilés R. Regulation of SERCA pumps expression in diabetes. Cell Calcium 2014; 56: 302-310
  • 15 Duncan JG. Mitochondrial dysfunction in diabetic cardiomyopathy. Biochim Biophys Acta 2011; 1813: 1351-1359
  • 16 Khanna S, Singh GB, Khullar M. Nitric oxide synthases and diabetic cardiomyopathy. Nitric Oxide 2014; 43: 29-34
  • 17 Tarquini R, Lazzeri C, Pala L. et al. The diabetic cardiomyopathy. Acta Diabetol 2011; 48: 173-181
  • 18 Rosa CM, Xavier NP, Henrique Campos D. et al. Diabetes mellitus activates fetal gene program and intensifies cardiac remodeling and oxidative stress in aged spontaneously hypertensive rats. Cardiovasc Diabetol 2013; 17 12: 152
  • 19 Boudina S, Abel ED. Diabetic cardiomyopathy, causes and effects. Rev Endocr Metab Disord 2010; 11: 31-39
  • 20 Watanabe K, Thandavarayan RA, Harima M. et al. Role of differential signaling pathways and oxidative stress in diabetic cardiomyopathy. Curr Cardiol Rev 2010; 6: 280-290
  • 21 Zhao CT, Wang M, Siu CW. et al. Myocardial dysfunction in patients with type 2 diabetesmellitus: role of endothelial progenitor cells and oxidative stress. Cardiovasc Diabetol 2012; 11: 147
  • 22 Hill MF. Diabetic Cardiomyopathy: Cardiac Changes, Pathophysiological Mechanisms, Biologic Markers, and the Available Therapeutic Armamentarium. In: Veselka J. (ed.). Cardiomyopathies – From Basic Research to Clinical Management: InTech. 2012. p 800
  • 23 Aragno M, Mastrocola R, Catalano MG. et al. Oxidative stress impairs skeletal muscle repair in diabetic rats. Diabetes 2004; 53: 1082-1088
  • 24 Wolff SP. Diabetes mellitus and free radicals. Free radicals, transition metals and oxidative stress in the aetiology of diabetes mellitus and complications. Br Med Bull 1993; 49: 642-652
  • 25 Mullarkey CJ, Edelstein D, Brownlee M. Free radical generation by early glycation products: a mechanism for accelerated atherogenesis in diabetes. Biochem Biophys Res Commun 1990; 173: 932-939
  • 26 Hunt JV, Smith CC, Wolff SP. Autooxidative glycosylation and possible involvement of peroxides and free radicals in LDL modification by glucose. Diabetes 1990; 39: 1420-1424
  • 27 Nielsen F, Mikkelsen BB, Nielsen JB. et al. Plasma malondialdehyde as biomarker for oxidative stress: reference interval and effects of life-style factors. Clin Chem 1997; 43: 1209-1214
  • 28 Fujii J, Iuchi Y, Matsuki S. et al. Cooperative function of antioxidant and redox systems against oxidative stress in male reproductive tissues. Asian J Androl 2003; 5: 231-242
  • 29 American Diabetes Association. Standards of medical care in diabetes-2012. Diabetes Care 2012; 34: S11-S61
  • 30 Marwick TH, Hordern MD, Miller T. et al. Exercise Training for Type 2 Diabetes Mellitus Impact on Cardiovascular Risk: A Scientific State-ment From the American Heart Association. Circulation 2009; 119: 3244-3262
  • 31 Eun-Sang 1 J, Chang-Ju K, Jun Heon P. et al. Duration-dependence of the effect of treadmill exercise on hyperactivity in attention deficit hyperactivity disorder rats. J Exerc Rehabil 2014; 10: 75-80
  • 32 Passey RB, Gillum RF, Fuller JB. et al. Evaluation and comparison of 10 glucose methods and the reference method recommended in the proposed product class standard. Clin Chem 1977; 23: 131-139
  • 33 Hales CN, Randle PJ. Immunoassay of insulin with insulin-antibody precipitate. Biochem J 1963; 88: 137-146
  • 34 Smith PK, Krohn RI, Hermanson GT. et al. Measurement of protein using bicinchoninic acid. Anal Biochem 1985; 150: 76-85
  • 35 Buage JA, Aust SD. Microsomal lipid peroxidation. Methods Enzymol 1978; 12: 302-310
  • 36 Mylorie AA, Collins H, Umbles C. et al. Erythrocyte superoxide dismutase activity and other parameters of copper status in rats ingesting lead acetate. Toxicol Appl Pharmacol 1986; 82: 512-520
  • 37 Lawrence RA, Burk RF. Glutathione peroxidase activity in selenium deficient rat liver. Biochem Biophys Res Commun 1976; 71: 952-958
  • 38 Aebi H. Catalase in vitro. Methods Enzymol 1974; 105: 121-126
  • 39 Hsu SM, Raine L, Fanger H. Use of avidin–biotin-peroxidase complex (ABC) in immunperoxidase techniques: a comparison between ABC and unlabeled antibody (PAP) procedures. J Histochem Cytochem 1980; 29: 577-580
  • 40 Grankvist K, Marklund SL, Taljedal LB. CuZn-superoxide dismutase, Mn-superoxide dismutase, catalase and glutathione peroxidase in pancreatic islets and other tissues in the mouse. Biochem J 1981; 199: 393-398
  • 41 Kanter M, Yoruk M, Koc A. et al. Effects of cadmium exposure on morphological aspects of pancreas, weights of fetus and placenta in streptozotocin-induced diabetic pregnant rats. Biol Trace Elem Res 2003; 93: 189-200
  • 42 Derouich M, Boutayeb A. The effect of physical exercise on the dynamics of glucose and insulin. J Biomech 2002; 35: 911-917
  • 43 Gul M, Laaksonen DE, Atalay M. et al. Effects of endurance training on tissue glutathione homeostasis and lipid peroxidation in streptozotocin-induced diabetic rats. Scand. J. Med. Sci. Sports. 2002; 12: 163-170
  • 44 Hammes HP, Martin S, Federlin K. et al. Aminoguanidine treatment inhibits the development of experimental diabetic retinopathy. Proc Natl Acad Sci USA 1991; 88: 11555-11558
  • 45 Sakurai T, Tsuchiya S. Superoxide production from nonenzymatically glycated protein. FEBS Lett 1988; 236: 406-410
  • 46 Hunt JV, Smith CC, Wolff SP. Autoxidative glycosylation and possible involvement of peroxides and free radicals in LDL modification by glucose. Diabetes 1990; 39: 1420-1424
  • 47 Matsuoka T, Kajimoto Y, Watada H. et al. Glycation-dependent, reactive oxygen species-mediated suppression of the insulin gene promoter activity in HIT cells. J Clin Invest 1997; 99: 144-150
  • 48 Panzram G. Mortality and survival in type 2 (non-insulin-dependent) diabetes mellitus. Diabetologia 1987; 30: 123-131
  • 49 Grossman E, Messerli FH. Diabetic and hypertensive heart disease. Ann Intern Med 1996; 125: 304-310
  • 50 Hsiao YC, Suzuki K, Abe H. et al. Ultrastructural alterations in cardiac muscle of diabetic BB Wistar rats. Virchows Arch A Pathol Anat Histopathol 1987; 411: 45-52
  • 51 Factor SM, Minase T, Sonnenblick EH. Clinical and morphological features of human hypertensive-diabetic cardiomyopathy. Am Heart J 1980; 99: 446-458
  • 52 Litwin SE, Raya TE, Anderson PG. et al. Abnormal cardiac function in the streptozotocin-diabetic rat. Changes in active and passive properties of the left ventricle. J Clin Invest 1990; 86: 481-488
  • 53 Searls YM, Smirnova IV, Fegley BR. et al. Exercise attenuates diabetes-induced ultrastructural changes in rat cardiac tissue. Medicine and science in sports and exercise 2004; 36: 1863-1870
  • 54 Boudina S, Abel ED. Diabetic cardiomyopathy revisited. Circulation 2007; 115: 3213-3223
  • 55 Poornima IG, Parikh P, Shannon RP. Diabetic cardiomyopathy: the search for a unifying hypothesis. Circ Res 2006; 98: 596-605
  • 56 Fiordaliso F, Li B, Latini R. et al. Myocyte death in streptozotocin-induced diabetes in rats is angiotensin II-dependent. Lab Invest 2000; 80: 513-527
  • 57 Fiordaliso F, De Angelis N, Bai A. et al. Effect of beta-adrenergic and renin-angiotensin system blockade on myocyte apoptosis and oxidative stress in diabetic hypertensive rats. Life Sci 2007; 81: 951-959
  • 58 Hamblin M, Friedman DB, Hill S. et al. Alterations in the diabetic myocardial proteome coupled with increased myocardial oxidative stress underlies diabetic cardiomyopathy. J Mol Cell Cardiol 2007; 42: 884-895
  • 59 Poornima IG, Parikh P, Shannon RP. Diabetic cardiomyopathy: the search for a unifying hypothesis. Circ Res 2006; 98: 596-605
  • 60 Westermann D, Rutschow S, Jäger S. et al. Contributions of inflammation and cardiac matrix metalloproteinase activity to cardiac failure in diabetic cardiomyopathy: the role of angiotensin type 1 receptor antagonism. Diabetes 2007; 56: 641-646
  • 61 Westermann D, Van Linthout S, Dhayat S. et al. Cardioprotective and anti-inflammatory effects of interleukin converting enzyme inhibition in experimental diabetic cardiomyopathy. Diabetes 2007; 56: 1834-1841
  • 62 Samadi A, Gaeini A, Hedayati M. et al. The effect of resistance exercise on oxidative stress in cardiac and skeletal muscle tissues of streptozotocin-induced diabetic rats. Journal of Basic and Clinical Pathophysiology 2013; 2: 28-33
  • 63 Eriksson UJ, Borg LAH. Protection by free oxygen radical scavenging enzymes against glucose-induced embryonic malformations in vitro. Diabetes 1991; 34: 325-331
  • 64 Gutteridge JMC. Lipid peroxidation and antioxidants as biomarkers of tissue damage. Clin Chem 1995; 41: 1819-1828
  • 65 Izuka Y, Sakurai E, Hikichi N. Effects of selenium on the serum glucose and insulin levels in diabetic rats. Nippon Yakurigaku Zassh 1992; 100: 151-154
  • 66 Kamalakkannan N, Stanely Mainzen Prince P. Effects of Aegle marmelos Correa (Bael) fruit extract on tissue antioxidants in streptozotocin diabetic rats. Indian J Exp Biol 2003; 41: 1285-1288
  • 67 Wang GG, Li W, Lu XH. et al. Taurine attenuates oxidative stress and alleviates cardiac failure in type I diabetic rats. Croat Med J 2013; 54: 171-179 10.3325/cmj.2013.54.171
  • 68 Montilla PL, Vargas JF, Tunez IF. et al. Oxidative stress in diabetic rats induced by streptozotocin: protective effects of melatonin. J Pineal Res 1998; 25: 94-100 10.1111/j.1600-079x.1998.tb00545.x
  • 69 Bao H, Chen L. Icariin reduces mitochondrial oxidative stress injury in diabetic rat hearts. Zhongguo Zhong Yao Za Zhi 2011; 36: 1503-1507 10.4268/cjcmm20111121