Yeungnam Univ J Med.  2021 Apr;38(2):83-94. 10.12701/yujm.2020.00563.

Can antioxidants be effective therapeutics for type 2 diabetes?

Affiliations
  • 1Department of Physiology and Smart-aging Convergence Research Center, Yeungnam University College of Medicine, Daegu, Korea

Abstract

The global obesity epidemic and the growing elderly population largely contribute to the increasing incidence of type 2 diabetes. Insulin resistance acts as a critical link between the present obesity pandemic and type 2 diabetes. Naturally occurring reactive oxygen species (ROS) regulate intracellular signaling and are kept in balance by the antioxidant system. However, the imbalance between ROS production and antioxidant capacity causes ROS accumulation and induces oxidative stress. Oxidative stress interrupts insulin-mediated intracellular signaling pathways, as supported by studies involving genetic modification of antioxidant enzymes in experimental rodents. In addition, a close association between oxidative stress and insulin resistance has been reported in numerous human studies. However, the controversial results with the use of antioxidants in type 2 diabetes raise the question of whether oxidative stress plays a critical role in insulin resistance. In this review article, we discuss the relevance of oxidative stress to insulin resistance based on genetically modified animal models and human trials.

Keyword

Antioxidants; Insulin resistance; Oxidative stress; Reactive oxygen species

Figure

  • Fig. 1. Intracellular reactive oxygen species (ROS) generation and the antioxidant scavenging system. ROS is produced from mitochondria, peroxisome, nicotinamide-adenine dinucleotide phosphate (NADPH) oxidase, and xanthine oxidase. Among these sources, the mitochondrial electron transport chain is the primary source for ROS production. Superoxide dismutase 2 (SOD2) catalyzes the conversion of superoxide anion (O2–) into hydrogen peroxide (H2O2) in the mitochondria. The H2O2 is then detoxified in the mitochondria or moves to the cytoplasm. Cytoplasmic superoxide anion is generated from NADPH oxidase and xanthine oxidase, and subsequently converted into H2O2 by superoxide dismutase 1 (SOD1). H2O2 is detoxified by glutathione (GSH)/glutaredoxin (GRx)/glutathione reductase (GR), catalase (CAT), peroxiredoxin (Prx), thioredoxin (Trx)/thioredoxin reductase (TrxR), and glutathione peroxidase (GPx). Oxidized methionine (Ox-Met) is reduced by methionine sulfoxide reductase (Msr) to methionine (Met). Nitric oxide (NO) reacts with superoxide anion to form peroxynitrite (ONOO–), which is detoxified by Prx, GSH, and GPx.

  • Fig. 2. Intracellular insulin signaling pathway in skeletal muscle. Binding of insulin to insulin receptors (IR) on the plasma membrane promotes tyrosine autophosphorylation at the IR, which in turn induces tyrosine phosphorylation of the IR substrate (IRS). IRS activates the downstream substrate phosphatidylinositol 3-kinases (PI3K)/protein kinase B (AKT) pathway, and the activated AKT leads to increased glucose uptake and glycogen synthesis by inducing phosphorylation of AKT substrate of 160 kDa (AS160) and glycogen synthase kinase 3 (GSK3), respectively. Activated AS160 increases glucose uptake by mediating the translocation of glucose transporter type 4 (GLUT4) from the cytoplasm to the plasma membrane. Intracellular glucose is used for adenosine triphosphate (ATP) generation and glycogen synthesis. Tyr, tyrosine; Ser, serine; PIP2, phosphatidylinositol 4,5-bisphosphate; PIP3, phosphatidylinositol (3,4,5)-triphosphate; PDK1, phosphatidylinositide-dependent protein kinase 1; mTORC2, mammalian target of rapamycin complex 2; FOXO1, forkhead box protein O1; GS, glycogen synthase; acetyl-CoA, acetyl coenzyme A; TCA, tricarboxylic acid cycle; ETC, electron transport chain; ROS, reactive oxygen species.

  • Fig. 3. Inhibition of the insulin signaling pathway by oxidative stress. Reactive oxygen species (ROS) interferes with insulin action by altering several substrates of the insulin signaling pathway. ROS activates serine/threonine kinases, including protein kinase C (PKC), c-Jun N-terminal kinase (JNK), and inhibitory κB kinase (IKK), which not only inhibit the activation of insulin receptor substrate (IRS) through serine phosphorylation but also induce inflammation by activating nuclear factor κB (NF-κB). In addition, ROS suppresses glucose absorption by degrading glucose transporter type 4 (GLUT4) in a casein kinase 2 (CK2)-dependent manner. Reactive nitrogen species (RNS) inhibits tyrosine phosphorylation of IRS and protein kinase B (AKT) activation by inducing nitration of tyrosine. Mitochondrial functional defects by ROS not only induce an explosive increase in oxidative stress but also suppress mitochondrial energy production, eventually leading to cell death. Tyr, tyrosine; Ser, serine; PIP2, phosphatidylinositol 4,5-bisphosphate; PIP3, phosphatidylinositol (3,4,5)-triphosphate; PI3K, phosphatidylinositol 3-kinases; PDK1, phosphatidylinositide-dependent protein kinase 1; mTORC2, mammalian target of rapamycin complex 2; FOXO1, forkhead box protein O1; GSK3, glycogen synthase kinase 3; AS160, AKT substrate of 160 kDa; acetyl-CoA, acetyl coenzyme A; mtDNA, mitochondrial DNA; TCA, tricarboxylic acid cycle; ETC, electron transport chain.


Reference

References

1. Abdali D, Samson SE, Grover AK. How effective are antioxidant supplements in obesity and diabetes? Med Princ Pract. 2015; 24:201–15.
Article
2. Forbes JM, Cooper ME. Mechanisms of diabetic complications. Physiol Rev. 2013; 93:137–88.
Article
3. Di Meo S, Iossa S, Venditti P. Skeletal muscle insulin resistance: role of mitochondria and other ROS sources. J Endocrinol. 2017; 233:R15–42.
Article
4. Facchini FS, Hua N, Abbasi F, Reaven GM. Insulin resistance as a predictor of age-related diseases. J Clin Endocrinol Metab. 2001; 86:3574–8.
Article
5. Rains JL, Jain SK. Oxidative stress, insulin signaling, and diabetes. Free Radic Biol Med. 2011; 50:567–75.
Article
6. Li R, Jia Z, Trush MA. Defining ROS in biology and medicine. React Oxyg Species (Apex). 2016; 1:9–21.
Article
7. He L, He T, Farrar S, Ji L, Liu T, Ma X. Antioxidants maintain cellular redox homeostasis by elimination of reactive oxygen species. Cell Physiol Biochem. 2017; 44:532–53.
Article
8. Trujillo M, Ferrer-Sueta G, Radi R. Peroxynitrite detoxification and its biologic implications. Antioxid Redox Signal. 2008; 10:1607–20.
Article
9. Cheung PY, Wang W, Schulz R. Glutathione protects against myocardial ischemia-reperfusion injury by detoxifying peroxynitrite. J Mol Cell Cardiol. 2000; 32:1669–78.
Article
10. Herrera R, Rosen OM. Autophosphorylation of the insulin receptor in vitro. Designation of phosphorylation sites and correlation with receptor kinase activation. J Biol Chem. 1986; 261:11980–5.
Article
11. Sun XJ, Rothenberg P, Kahn CR, Backer JM, Araki E, Wilden PA, et al. Structure of the insulin receptor substrate IRS-1 defines a unique signal transduction protein. Nature. 1991; 352:73–7.
Article
12. Hanke S, Mann M. The phosphotyrosine interactome of the insulin receptor family and its substrates IRS-1 and IRS-2. Mol Cell Proteomics. 2009; 8:519–34.
Article
13. Taniguchi CM, Kondo T, Sajan M, Luo J, Bronson R, Asano T, et al. Divergent regulation of hepatic glucose and lipid metabolism by phosphoinositide 3-kinase via Akt and PKClambda/zeta. Cell Metab. 2006; 3:343–53.
14. Alessi DR, James SR, Downes CP, Holmes AB, Gaffney PR, Reese CB, et al. Characterization of a 3-phosphoinositide-dependent protein kinase which phosphorylates and activates protein kinase Balpha. Curr Biol. 1997; 7:261–9.
15. Sarbassov DD, Guertin DA, Ali SM, Sabatini DM. Phosphorylation and regulation of Akt/PKB by the rictor-mTOR complex. Science. 2005; 307:1098–101.
Article
16. Manning BD, Toker A. AKT/PKB signaling: navigating the network. Cell. 2017; 169:381–405.
Article
17. Imazu M, Strickland WG, Chrisman TD, Exton JH. Phosphorylation and inactivation of liver glycogen synthase by liver protein kinases. J Biol Chem. 1984; 259:1813–21.
Article
18. Kwon HS, Huang B, Unterman TG, Harris RA. Protein kinase B-alpha inhibits human pyruvate dehydrogenase kinase-4 gene induction by dexamethasone through inactivation of FOXO transcription factors. Diabetes. 2004; 53:899–910.
Article
19. Sano H, Kane S, Sano E, Mîinea CP, Asara JM, Lane WS, et al. Insulin-stimulated phosphorylation of a Rab GTPase-activating protein regulates GLUT4 translocation. J Biol Chem. 2003; 278:14599–602.
Article
20. Cerf ME. Beta cell dysfunction and insulin resistance. Front Endocrinol (Lausanne). 2013; 4:37.
Article
21. Kashyap SR, Belfort R, Berria R, Suraamornkul S, Pratipranawatr T, Finlayson J, et al. Discordant effects of a chronic physiological increase in plasma FFA on insulin signaling in healthy subjects with or without a family history of type 2 diabetes. Am J Physiol Endocrinol Metab. 2004; 287:E357–46.
Article
22. Santomauro AT, Boden G, Silva ME, Rocha DM, Santos RF, Ursich MJ, et al. Overnight lowering of free fatty acids with Acipimox improves insulin resistance and glucose tolerance in obese diabetic and nondiabetic subjects. Diabetes. 1999; 48:1836–41.
Article
23. Itani SI, Ruderman NB, Schmieder F, Boden G. Lipid-induced insulin resistance in human muscle is associated with changes in diacylglycerol, protein kinase C, and IkappaB-alpha. Diabetes. 2002; 51:2005–11.
24. Holland WL, Bikman BT, Wang LP, Yuguang G, Sargent KM, Bulchand S, et al. Lipid-induced insulin resistance mediated by the proinflammatory receptor TLR4 requires saturated fatty acid-induced ceramide biosynthesis in mice. J Clin Invest. 2011; 121:1858–70.
Article
25. Oakes ND, Bell KS, Furler SM, Camilleri S, Saha AK, Ruderman NB, et al. Diet-induced muscle insulin resistance in rats is ameliorated by acute dietary lipid withdrawal or a single bout of exercise: parallel relationship between insulin stimulation of glucose uptake and suppression of long-chain fatty acyl-CoA. Diabetes. 1997; 46:2022–8.
Article
26. Arkan MC, Hevener AL, Greten FR, Maeda S, Li ZW, Long JM, et al. IKK-beta links inflammation to obesity-induced insulin resistance. Nat Med. 2005; 11:191–8.
Article
27. Hotamisligil GS, Peraldi P, Budavari A, Ellis R, White MF, Spiegelman BM. IRS-1-mediated inhibition of insulin receptor tyrosine kinase activity in TNF-alpha- and obesity-induced insulin resistance. Science. 1996; 271:665–8.
Article
28. Barazzoni R, Zanetti M, Gortan Cappellari G, Semolic A, Boschelle M, Codarin E, et al. Fatty acids acutely enhance insulin-induced oxidative stress and cause insulin resistance by increasing mitochondrial reactive oxygen species (ROS) generation and nuclear factor-κB inhibitor (IκB)-nuclear factor-κB (NFκB) activation in rat muscle, in the absence of mitochondrial dysfunction. Diabetologia. 2012; 55:773–82.
Article
29. Satapati S, Kucejova B, Duarte JA, Fletcher JA, Reynolds L, Sunny NE, et al. Mitochondrial metabolism mediates oxidative stress and inflammation in fatty liver. J Clin Invest. 2015; 125:4447–62.
Article
30. Paglialunga S, Ludzki A, Root-McCaig J, Holloway GP. In adipose tissue, increased mitochondrial emission of reactive oxygen species is important for short-term high-fat diet-induced insulin resistance in mice. Diabetologia. 2015; 58:1071–80.
Article
31. Chen XH, Zhao YP, Xue M, Ji CB, Gao CL, Zhu JG, et al. TNF-alpha induces mitochondrial dysfunction in 3T3-L1 adipocytes. Mol Cell Endocrinol. 2010; 328:63–9.
32. Imoto K, Kukidome D, Nishikawa T, Matsuhisa T, Sonoda K, Fujisawa K, et al. Impact of mitochondrial reactive oxygen species and apoptosis signal-regulating kinase 1 on insulin signaling. Diabetes. 2006; 55:1197–204.
Article
33. Ma J, Nakagawa Y, Kojima I, Shibata H. Prolonged insulin stimulation down-regulates GLUT4 through oxidative stress-mediated retromer inhibition by a protein kinase CK2-dependent mechanism in 3T3-L1 adipocytes. J Biol Chem. 2014; 289:133–42.
Article
34. Nomiyama T, Igarashi Y, Taka H, Mineki R, Uchida T, Ogihara T, et al. Reduction of insulin-stimulated glucose uptake by peroxynitrite is concurrent with tyrosine nitration of insulin receptor substrate-1. Biochem Biophys Res Commun. 2004; 320:639–47.
Article
35. Zou MH, Hou XY, Shi CM, Nagata D, Walsh K, Cohen RA. Modulation by peroxynitrite of Akt- and AMP-activated kinase-dependent Ser1179 phosphorylation of endothelial nitric oxide synthase. J Biol Chem. 2002; 277:32552–7.
Article
36. Bonnard C, Durand A, Peyrol S, Chanseaume E, Chauvin MA, Morio B, et al. Mitochondrial dysfunction results from oxidative stress in the skeletal muscle of diet-induced insulin-resistant mice. J Clin Invest. 2008; 118:789–800.
Article
37. Hiona A, Sanz A, Kujoth GC, Pamplona R, Seo AY, Hofer T, et al. Mitochondrial DNA mutations induce mitochondrial dysfunction, apoptosis and sarcopenia in skeletal muscle of mitochondrial DNA mutator mice. PLoS One. 2010; 5:e11468.
Article
38. Bhatti JS, Bhatti GK, Reddy PH. Mitochondrial dysfunction and oxidative stress in metabolic disorders - a step towards mitochondria based therapeutic strategies. Biochim Biophys Acta Mol Basis Dis. 2017; 1863:1066–77.
Article
39. Shin MG, Cha HN, Park S, Kim YW, Kim JY, Park SY. Selenoprotein W deficiency does not affect oxidative stress and insulin sensitivity in the skeletal muscle of high-fat diet-fed obese mice. Am J Physiol Cell Physiol. 2019; 317:C1172–82.
Article
40. Heo JY, Cha HN, Kim KY, Lee E, Kim SJ, Kim YW, et al. Methionine sulfoxide reductase B1 deficiency does not increase high-fat diet-induced insulin resistance in mice. Free Radic Res. 2017; 51:24–37.
Article
41. Muscogiuri G, Salmon AB, Aguayo-Mazzucato C, Li M, Balas B, Guardado-Mendoza R, et al. Genetic disruption of SOD1 gene causes glucose intolerance and impairs β-cell function. Diabetes. 2013; 62:4201–7.
Article
42. Liu Y, Qi W, Richardson A, Van Remmen H, Ikeno Y, Salmon AB. Oxidative damage associated with obesity is prevented by overexpression of CuZn- or Mn-superoxide dismutase. Biochem Biophys Res Commun. 2013; 438:78–83.
Article
43. Kang L, Dai C, Lustig ME, Bonner JS, Mayes WH, Mokshagundam S, et al. Heterozygous SOD2 deletion impairs glucose-stimulated insulin secretion, but not insulin action, in high-fat-fed mice. Diabetes. 2014; 63:3699–710.
Article
44. Lark DS, Kang L, Lustig ME, Bonner JS, James FD, Neufer PD, et al. Enhanced mitochondrial superoxide scavenging does not improve muscle insulin action in the high fat-fed mouse. PLoS One. 2015; 10:e0126732.
Article
45. Boden MJ, Brandon AE, Tid-Ang JD, Preston E, Wilks D, Stuart E, et al. Overexpression of manganese superoxide dismutase ameliorates high-fat diet-induced insulin resistance in rat skeletal muscle. Am J Physiol Endocrinol Metab. 2012; 303:E798–805.
Article
46. Park YS, Uddin MJ, Piao L, Hwang I, Lee JH, Ha H. Novel role of endogenous catalase in macrophage polarization in adipose tissue. Mediators Inflamm. 2016; 2016:8675905.
Article
47. Piao L, Dorotea D, Jiang S, Koh EH, Oh GT, Ha H. Impaired peroxisomal fitness in obese mice, a vicious cycle exacerbating adipocyte dysfunction via oxidative stress. Antioxid Redox Signal. 2019; 31:1339–51.
48. Amos DL, Robinson T, Massie MB, Cook C, Hoffsted A, Crain C, et al. Catalase overexpression modulates metabolic parameters in a new ‘stress-less’ leptin-deficient mouse model. Biochim Biophys Acta Mol Basis Dis. 2017; 1863:2293–306.
Article
49. Kang L, Lustig ME, Bonner JS, Lee-Young RS, Mayes WH, James FD, et al. Mitochondrial antioxidative capacity regulates muscle glucose uptake in the conscious mouse: effect of exercise and diet. J Appl Physiol (1985). 2012; 113:1173–83.
Article
50. Lee HY, Lee JS, Alves T, Ladiges W, Rabinovitch PS, Jurczak MJ, et al. Mitochondrial-targeted catalase protects against high-fat diet-induced muscle insulin resistance by decreasing intramuscular lipid accumulation. Diabetes. 2017; 66:2072–81.
Article
51. McClung JP, Roneker CA, Mu W, Lisk DJ, Langlais P, Liu F, et al. Development of insulin resistance and obesity in mice overexpressing cellular glutathione peroxidase. Proc Natl Acad Sci U S A. 2004; 101:8852–7.
Article
52. Loh K, Deng H, Fukushima A, Cai X, Boivin B, Galic S, et al. Reactive oxygen species enhance insulin sensitivity. Cell Metab. 2009; 10:260–72.
Article
53. Merry TL, Tran M, Dodd GT, Mangiafico SP, Wiede F, Kaur S, et al. Hepatocyte glutathione peroxidase-1 deficiency improves hepatic glucose metabolism and decreases steatohepatitis in mice. Diabetologia. 2016; 59:2632–44.
Article
54. Kim HR, Choi EJ, Kie JH, Lee JH, Seoh JY. Deficiency of glutathione peroxidase-1 and catalase attenuated diet-induced obesity and associated metabolic disorders. Acta Diabetol. 2020; 57:151–61.
Article
55. Wohua Z, Weiming X. Glutaredoxin 2 (GRX2) deficiency exacerbates high fat diet (HFD)-induced insulin resistance, inflammation and mitochondrial dysfunction in brain injury: a mechanism involving GSK-3β. Biomed Pharmacother. 2019; 118:108940.
Article
56. Cha HN, Park S, Dan Y, Kim JR, Park SY. Peroxiredoxin2 deficiency aggravates aging-induced insulin resistance and declines muscle strength. J Gerontol A Biol Sci Med Sci. 2019; 74:147–54.
Article
57. Kim JH, Park SJ, Chae U, Seong J, Lee HS, Lee SR, et al. Peroxiredoxin 2 mediates insulin sensitivity of skeletal muscles through regulation of protein tyrosine phosphatase oxidation. Int J Biochem Cell Biol. 2018; 99:80–90.
Article
58. Kim JH, Cha HN, Kim YW, Park SY. Peroxiredoxin 2 deficiency does not affect insulin resistance and oxidative stress in high-fat diet-fed obese mice. Arch Physiol Biochem. 2020; Mar. 6. [Epub]. Arch Physiol Biochem https://doi.org/10.1080/13813455.2020.1733026.
Article
59. Huh JY, Kim Y, Jeong J, Park J, Kim I, Huh KH, et al. Peroxiredoxin 3 is a key molecule regulating adipocyte oxidative stress, mitochondrial biogenesis, and adipokine expression. Antioxid Redox Signal. 2012; 16:229–43.
Article
60. Chen L, Na R, Gu M, Salmon AB, Liu Y, Liang H, et al. Reduction of mitochondrial H2O2 by overexpressing peroxiredoxin 3 improves glucose tolerance in mice. Aging Cell. 2008; 7:866–78.
61. Nabeshima A, Yamada S, Guo X, Tanimoto A, Wang KY, Shimajiri S, et al. Peroxiredoxin 4 protects against nonalcoholic steatohepatitis and type 2 diabetes in a nongenetic mouse model. Antioxid Redox Signal. 2013; 19:1983–98.
Article
62. Pacifici F, Arriga R, Sorice GP, Capuani B, Scioli MG, Pastore D, et al. Peroxiredoxin 6, a novel player in the pathogenesis of diabetes. Diabetes. 2014; 63:3210–20.
Article
63. Styskal J, Nwagwu FA, Watkins YN, Liang H, Richardson A, Musi N, et al. Methionine sulfoxide reductase A affects insulin resistance by protecting insulin receptor function. Free Radic Biol Med. 2013; 56:123–32.
64. Hunnicut J, Liu Y, Richardson A, Salmon AB. MsrA overexpression targeted to the mitochondria, but not cytosol, preserves insulin sensitivity in diet-induced obese mice. PLoS One. 2015; 10:e0139844.
Article
65. Nishikawa T, Araki E. Impact of mitochondrial ROS production in the pathogenesis of diabetes mellitus and its complications. Antioxid Redox Signal. 2007; 9:343–53.
Article
66. Li Y, Soos TJ, Li X, Wu J, Degennaro M, Sun X, et al. Protein kinase C theta inhibits insulin signaling by phosphorylating IRS1 at Ser(1101). J Biol Chem. 2004; 279:45304–7.
67. Ju TJ, Kwon WY, Kim YW, Kim JY, Kim YD, Lee IK, et al. Hemin improves insulin sensitivity in skeletal muscle in high fat-fed mice. J Pharmacol Sci. 2014; 126:115–25.
Article
68. Kim BS, Cha HN, Kim YW, Kim JY, Dan JM, Park SY. Inhibition of lipid infusion-induced skeletal muscle insulin resistance by cotreatment with tempol and glutathione in mice. J Pharmacol Sci. 2009; 110:370–80.
Article
69. Gao M, Zhao Z, Lv P, Li Y, Gao J, Zhang M, et al. Quantitative combination of natural anti-oxidants prevents metabolic syndrome by reducing oxidative stress. Redox Biol. 2015; 6:206–17.
Article
70. Lebel M, Massip L, Garand C, Thorin E. Ascorbate improves metabolic abnormalities in Wrn mutant mice but not the free radical scavenger catechin. Ann N Y Acad Sci. 2010; 1197:40–4.
Article
71. Telci A, Cakatay U, Kayali R, Erdoğan C, Orhan Y, Sivas A, et al. Oxidative protein damage in plasma of type 2 diabetic patients. Horm Metab Res. 2000; 32:40–3.
Article
72. Piwowar A, Knapik-Kordecka M, Warwas M. Markers of oxidative protein damage in plasma and urine of type 2 diabetic patients. Br J Biomed Sci. 2009; 66:194–9.
Article
73. Atli T, Keven K, Avci A, Kutlay S, Turkcapar N, Varli M, et al. Oxidative stress and antioxidant status in elderly diabetes mellitus and glucose intolerance patients. Arch Gerontol Geriatr. 2004; 39:269–75.
Article
74. Song F, Jia W, Yao Y, Hu Y, Lei L, Lin J, et al. Oxidative stress, antioxidant status and DNA damage in patients with impaired glucose regulation and newly diagnosed type 2 diabetes. Clin Sci (Lond). 2007; 112:599–606.
Article
75. Konukoglu D, Turhan MS, Ercan M, Serin O. Relationship between plasma leptin and zinc levels and the effect of insulin and oxidative stress on leptin levels in obese diabetic patients. J Nutr Biochem. 2004; 15:757–60.
Article
76. Ceriello A, Falleti E, Bortolotti N, Motz E, Cavarape A, Russo A, et al. Increased circulating intercellular adhesion molecule-1 levels in type II diabetic patients: the possible role of metabolic control and oxidative stress. Metabolism. 1996; 45:498–501.
Article
77. Dong QY, Cui Y, Chen L, Song J, Sun L. Urinary 8-hydroxydeoxyguanosine levels in diabetic retinopathy patients. Eur J Ophthalmol. 2008; 18:94–8.
Article
78. Cakatay U. Protein oxidation parameters in type 2 diabetic patients with good and poor glycaemic control. Diabetes Metab. 2005; 31:551–7.
Article
79. Cominacini L, Fratta Pasini A, Garbin U, Campagnola M, Davoli A, Rigoni A, et al. E-selectin plasma concentration is influenced by glycaemic control in NIDDM patients: possible role of oxidative stress. Diabetologia. 1997; 40:584–9.
Article
80. Suzuki S, Hinokio Y, Komatu K, Ohtomo M, Onoda M, Hirai S, et al. Oxidative damage to mitochondrial DNA and its relationship to diabetic complications. Diabetes Res Clin Pract. 1999; 45:161–8.
Article
81. Ingram KH, Hill H, Moellering DR, Hill BG, Lara-Castro C, Newcomer B, et al. Skeletal muscle lipid peroxidation and insulin resistance in humans. J Clin Endocrinol Metab. 2012; 97:E1182–6.
Article
82. Torres SH, De Sanctis JB, de L Briceño M, Hernández N, Finol HJ. Inflammation and nitric oxide production in skeletal muscle of type 2 diabetic patients. J Endocrinol. 2004; 181:419–27.
Article
83. Akkuş I, Kalak S, Vural H, Caglayan O, Menekşe E, Can G, et al. Leukocyte lipid peroxidation, superoxide dismutase, glutathione peroxidase and serum and leukocyte vitamin C levels of patients with type II diabetes mellitus. Clin Chim Acta. 1996; 244:221–7.
Article
84. Lin H, Ye S, Xu J, Wang W. The alpha-lipoic acid decreases urinary podocalyxin excretion in type 2 diabetics by inhibiting oxidative stress in vivo. J Diabetes Complications. 2015; 29:64–7.
Article
85. Furukawa S, Fujita T, Shimabukuro M, Iwaki M, Yamada Y, Nakajima Y, et al. Increased oxidative stress in obesity and its impact on metabolic syndrome. J Clin Invest. 2004; 114:1752–61.
Article
86. Anderson EJ, Lustig ME, Boyle KE, Woodlief TL, Kane DA, Lin CT, et al. Mitochondrial H2O2 emission and cellular redox state link excess fat intake to insulin resistance in both rodents and humans. J Clin Invest. 2009; 119:573–81.
Article
87. Konopka AR, Asante A, Lanza IR, Robinson MM, Johnson ML, Dalla Man C, et al. Defects in mitochondrial efficiency and H2O2 emissions in obese women are restored to a lean phenotype with aerobic exercise training. Diabetes. 2015; 64:2104–15.
Article
88. Tushuizen ME, Nieuwland R, Scheffer PG, Sturk A, Heine RJ, Diamant M. Two consecutive high-fat meals affect endothelial-dependent vasodilation, oxidative stress and cellular microparticles in healthy men. J Thromb Haemost. 2006; 4:1003–10.
Article
89. Samocha-Bonet D, Campbell LV, Mori TA, Croft KD, Greenfield JR, Turner N, et al. Overfeeding reduces insulin sensitivity and increases oxidative stress, without altering markers of mitochondrial content and function in humans. PLoS One. 2012; 7:e36320.
Article
90. Reitman A, Friedrich I, Ben-Amotz A, Levy Y. Low plasma antioxidants and normal plasma B vitamins and homocysteine in patients with severe obesity. Isr Med Assoc J. 2002; 4:590–3.
91. Hasegawa G, Yamamoto Y, Zhi JG, Tanino Y, Yamasaki M, Yano M, et al. Daily profile of plasma %CoQ10 level, a biomarker of oxidative stress, in patients with diabetes manifesting postprandial hyperglycaemia. Acta Diabetol. 2005; 42:179–81.
Article
92. van der Schaft N, Schoufour JD, Nano J, Kiefte-de Jong JC, Muka T, Sijbrands EJ, et al. Dietary antioxidant capacity and risk of type 2 diabetes mellitus, prediabetes and insulin resistance: the Rotterdam Study. Eur J Epidemiol. 2019; 34:853–61.
Article
93. Mancini FR, Affret A, Dow C, Balkau B, Bonnet F, Boutron-Ruault MC, et al. Dietary antioxidant capacity and risk of type 2 diabetes in the large prospective E3N-EPIC cohort. Diabetologia. 2018; 61:308–16.
Article
94. Kataja-Tuomola MK, Kontto JP, Männistö S, Albanes D, Virtamo JR. Effect of alpha-tocopherol and beta-carotene supplementation on macrovascular complications and total mortality from diabetes: results of the ATBC Study. Ann Med. 2010; 42:178–86.
Article
95. Sluijs I, Cadier E, Beulens JW, van der A DL, Spijkerman AM, van der Schouw YT. Dietary intake of carotenoids and risk of type 2 diabetes. Nutr Metab Cardiovasc Dis. 2015; 25:376–81.
Article
96. Eshak ES, Iso H, Muraki I, Tamakoshi A. Among the water-soluble vitamins, dietary intakes of vitamins C, B2 and folate are associated with the reduced risk of diabetes in Japanese women but not men. Br J Nutr. 2019; 121:1357–64.
Article
97. Seyyedebrahimi S, Khodabandehloo H, Nasli Esfahani E, Meshkani R. The effects of resveratrol on markers of oxidative stress in patients with type 2 diabetes: a randomized, double-blind, placebo-controlled clinical trial. Acta Diabetol. 2018; 55:341–53.
Article
98. Choi SW, Ho CK. Antioxidant properties of drugs used in type 2 diabetes management: could they contribute to, confound or conceal effects of antioxidant therapy? Redox Rep. 2018; 23:1–24.
Article
99. Dao VT, Casas AI, Maghzal GJ, Seredenina T, Kaludercic N, Robledinos-Anton N, et al. Pharmacology and clinical drug candidates in redox medicine. Antioxid Redox Signal. 2015; 23:1113–29.
Article
100. Bonetta R. Potential therapeutic applications of MnSODs and SOD-mimetics. Chemistry. 2018; 24:5032–41.
Article
Full Text Links
  • YUJM
Actions
Cited
CITED
export Copy
Close
Share
  • Twitter
  • Facebook
Similar articles
Copyright © 2024 by Korean Association of Medical Journal Editors. All rights reserved.     E-mail: koreamed@kamje.or.kr