Open access

Oxidative Stress and Diabetic Complications: The Role of Antioxidant Vitamins and Flavonoids

Written By

Omolola R. Ayepola, Nicole L. Brooks and Oluwafemi O. Oguntibeju

Submitted: 16 September 2013 Published: 05 February 2014

DOI: 10.5772/57282

From the Edited Volume

Antioxidant-Antidiabetic Agents and Human Health

Edited by Oluwafemi Oguntibeju

Chapter metrics overview

4,297 Chapter Downloads

View Full Metrics

1. Introduction

Diabetes mellitus is a group of disorders of multiple aetiologies resulting from a defect in insulin secretion, insulin action, or both. Insulin deficiency in turn leads to chronic hyperglycemia (very high blood glucose levels) with disturbances in carbohydrate, fat and protein metabolism [1]. The two major types of diabetes mellitus (DM) are insulin dependent (IDDM) - type 1 and non -insulin dependent (NIDDM) -type 2. Type 1 DM is characterized by a specific destruction of the pancreatic β cells commonly associated with immune-mediated damage [2]. Individuals with type 2 DM display a gradual change in glucose homeostasis due to insulin resistance and/or decreased insulin secretion [3].

Sustained hyperglycemia leads to the progressive development of long-term microvascular and macrovascular complications which causes morbidity and mortality among those affected [4, 5]. Although glycemic control has long been the mainstay for preventing the progression of diabetic complications, there is far less evidence that these interventions reverse diabetic complications [6]. Also, limitations in intensive glycemic treatment such as difficulty in achieving and/or maintaining tight glycemic control [7], incidence of hypoglycemia and increased mortality [8, 9] suggest an urgent need for alternative and/or complementary therapies to this disorder.

Hyperglycemia-induced oxidative stress is now recognized as the driving force for the development of diabetic complications [10]. Oxidative stress in diabetes results in stimulation of the polyol pathway, formation of advanced glycation end products (AGE), activation of protein kinase C (PKC) and subsequent formation of reactive oxygen radicals [11, 12]. Hyperglycemia, not only generates more reactive oxygen species (ROS), but also attenuates antioxidative mechanisms by scavenging enzymes and substances [13].

Advertisement

2. The complications of diabetes mellitus (DM)

The injurious effects of hyperglycemia are separated into microvascular (involving small vessels such as capillaries) and macrovascular complications (involving large vessels, such as arteries and veins). Microvascular complications include diabetic nephropathy, neuropathy and retinopathy while macrovascular complications include coronary artery disease, peripheral arterial disease and stroke [5].

Diabetic nephropathy is a major cause of end-stage renal disease worldwide. It is a progressive decline in the glomerular filtration rate, characterized by glomerular hyperfiltration, glomerular and tubular epithelial hypertrophy, increased urinary albumin excretion, increased basement membrane thickness and mesangial expansion with the accumulation of extracellular matrix proteins (ECM) [14]. Alteration of the permeability characteristics of the glomerular capillary wall manifests clinically as abnormal albuminuria [15]. Microalbuminuria progresses to end-stage renal disease through a number of stages including normoalbuminuria, microalbuminuria and macroalbuminuria [16].

Diabetic retinopathy results from the damage of the small vasculature of the retina, multi cellular and the light sensitive tissue at the back of the eye. It is a major cause of visual impairment worldwide [17, 18]. The retina capillaries are lined with endothelial cells responsible for maintaining the blood retinal barrier, and are surrounded by smooth muscle cells, pericytes, which provide tone to the vessels [18]. The vascular lesions that are identified at the early stage of diabetic retinopathy include pericytes disappearance from capillaries resulting in pericyte ghosts, obliteration of capillaries and small arterioles, gradual thickening of vascular basement membrane, increased permeability of endothelial cells, and formation of microaneurysms (i.e. weakening of vessel walls that results in the projection of a balloonlike sac), vessel leakage, exudate, and hemorrhage [19, 20].

Neuropathies are characterized by a progressive loss of nerve fiber function. A widely accepted definition of diabetic neuropathy is “the presence of symptoms and/or signs of peripheral nerve dysfunction in people with mellitus after exclusion of other causes” [21]. In the peripheral nervous system, diabetes causes a progressive deterioration of sensory nerves and damage to motor nerves [22]. Diabetic neuropathy is ultimately the leading cause of lower extremity amputation [23]. Peripheral neuropathy is thought to develop because of cellular damage to endothelial cells, affecting nerve blood flow and also damage to the neurons affecting conductivity of impulses [23]. Signs and symptoms of diabetic neuropathy include decrease or no sweating, numbness, or tingling, and some sort of burning sensation, weakness and loss of reflexes [24].

Both type I and type II diabetes are powerful and independent risk factors for coronary artery disease (CAD), stroke, and peripheral arterial disease [25, 26, 27]. Diabetics have a 2- to 4-fold higher risk for cardiovascular events [28] and nearly 80% of diabetes-associated deaths are caused by cardiovascular disease (CVD) [29]. Atherosclerosis, (excessive accumulation of lipids, cholesterol, inflammatory cells, and connective tissue in the vessel wall) accounts for more than 80% of the CVD-associated death and disability [30, 31]. Formation of atherosclerotic plaques can result in occlusion of vessel lumen and a rapid cessation in blood flow to target tissue [32]. Hyperglycemia, increased free fatty acids, and insulin resistance induce a large number of alterations at the cellular level that contribute to vascular dysfunction and accelerate the atherosclerotic process. These include increased oxidative stress, decreased bioavailability of NO, disturbances of intracellular signal transduction and increased production of several prothrombotic factors [32, 33].

Advertisement

3. Role of oxidative stress in diabetic complications

Reactive oxygen species (ROS) and reactive nitrogen species (RNS) are the terms collectively describing free radicals and other non-radical reactive derivatives also called oxidants. Biological free radicals are highly unstable molecules which are products of normal cellular metabolism. They have electrons available to react with various organic substrates such as lipids, proteins and deoxyribonucleic acid (DNA). Free radicals are well recognized for playing a dual role as both deleterious and beneficial species, since they can be either harmful or beneficial to living systems [34]. At low or moderate levels free radicals (ROS and RNS) exerts beneficial effects such as defence against infectious agents, induction of a mitogenic response and the maturation process of cellular structures [35-37]. ROS include superoxide anion (O2 .-), hydroxyl (.OH), hydrogen peroxide (H2O2) and hypochlorous acid (HOCl) while RNS include nitric oxide (.NO), nitrogen dioxide (NO2 .-) and peroxynitrite (OONO) [38, 39]. High concentrations of free radicals on the other hand result in deleterious processes that can damage cell structures due to oxidative stress [40, 41].

Free radicals produced under physiological conditions are maintained at steady state levels by endogenous or exogenous antioxidants (externally supplied through foods or supplements) which act as free radical scavengers. However, oxidative stress occurs when the production of free radicals overwhelms the detoxification capacity of cellular antioxidant system causing biological damage [42-44]. The endogenous antioxidants (Table 1) comprise of the enzymatic antioxidants such as superoxide dismutase (SOD), glutathione peroxidase (GPx), glutathione reductase (GR), catalase (CAT), and non-enzymatic antioxidants including glutathione (GSH), α lipoic acid, vitamins C and E [39, 45, 46]. On the other hand, the exogenous antioxidants include micronutrients and other exogenously administered compounds such as vitamin E, vitamin C, trace metals (selenium, manganese, zinc), carotenoids and flavonoids [39, 44, 47].

Antioxidants Cellular location Role Reference
Enzymatic Antioxidants
(A) Catalase Peroxisomes Decomposition of H2O2to water and oxygen [48]
(B) Glutathione peroxidase Cytoplasm, mitochondria, and nucleus Detoxifies H2O2 and lipid peroxides with simultaneous oxidation of GSH and generation of GSSG [49]
(C) Glutathione reductase Cytoplasm, mitochondria, and nucleus Recycles Glutathione disulfide back to glutathione using the cofactor NADPH [50]
(D) Superoxide dismutase Cytoplasm, nucleus lysosomes, mitochondria Conversion of superoxide radical to H2O2 [51]
Non enzymatic antioxidants
(A) GSH Cytoplasm, mitochondria and nucleus Acts as a cofactor for antioxidant enzymes (GPx, GST), regenerates other antioxidants such as Vitamins C and E to their active forms [52]
(B) Vitamin-E Membrane Directly scavenge singlet oxygen, peroxyl and superoxide radicals , protects against peroxidation of membrane lipids [34]
(C) Vitamin-C Cytosol Acts synergistically with vitamin E to terminate radical inducedlipid peroxidation [34, 53]
(D) α-Lipoic acid Cell membrane and cytoplasm Increases glutathione and vitamin C levels [54]

Table 1.

Role of antioxidants in the protection against free radical damage

Numerous experimental evidences have highlighted a direct link between oxidative stress and diabetes through the measurement of oxidative stress biomarkers in both diabetic patient and rodents. As shown in Table 2, a hyperglycemic state can lead to an increase in the levels of oxidative DNA damage markers such as 8-hydroxy-2’-deoxyguanosine (8-OHdG) and 8-oxo-7, 8-dihydro-2'-deoxyguanosine (8-oxodG); lipid-peroxidation products measured as thiobarbituric acid-reactive substances (TBARS); protein oxidation products such as nitrotyrosine and carbonyl levels and also lower the activity of antioxidant enzymes. Cell culture studies using pancreatic beta cells, aortic smooth muscle cells and endothelial cells have also provided evidence for an increase in ROS production in diabetes [55, 56].

Due to their ability to directly oxidize and damage DNA, proteins, and lipids, free radicals are believed to play a key role in the onset and progression of late-diabetic complications [57]. In the absence of an appropriate condensation by antioxidant defense network, increased oxidative stress leads to activation of stress-sensitive intracellular signaling pathways and the formation of gene products that cause cellular damage and contribute to late diabetic complications [58-61].

Antioxidants and macromolecules Evidence of oxidative stress Target tissue/organ References
Animals Enzymatic antioxidants SOD,CAT,GR, GPX Liver, Pancreas, Liver, kidney [62-64]
Non-enzymatic antioxidant Vit E and C Liver, kidney [64]
GSH /GSSG,GSH Kidney, hippocampus.
Retina,
Heart
[65]
[66]
[67]
[68]
Lipids TBARS, lipid peroxides, MDA Kidney [69-70]
DNA 8-OHdG, 8-OHG Plasma, Liver, Kidney [71-72]
Protein Nitrotyrosine kidney
Retina
[73]
[67]
Reactive oxygen species ROS Hippocampus [66]
Humans Enzymatic antioxidants SOD,CAT, GPX Erythrocyte [74]
Non enzymatic antioxidants GSH Erythrocyte [75]
Lipid F2-Isoprostanes
MDA
Urine
Erythrocyte
[76]
[77]
DNA 8-OHdG Urine [78-79]
Protein Nitrotyrosine
Protein carbonyl
Plasma [80, 81]

Table 2.

Experimental evidence supporting the involvement of oxidative stress

Advertisement

4. Pathways of free radical generation in diabetes mellitus and its associated complications

In diabetes, ROS is thought to be generated through increased polyol pathway [82], increased formation of advanced-glycation end products (AGEs) [83] and protein kinase C (PKC) activation [84].

4.1. Aldose reductase pathway and ROS generation

Aldose reductase is the rate limiting enzyme of the polyol pathway. The nicotinamide adenine dinucleotide phosphate (NAD(P)H)-requiring aldose reductase, catalyses the reduction of glucose to sorbitol followed by the oxidation of sorbitol to fructose by NAD+ dependent sorbitol dehydrogenase. At normal blood glucose concentration (5.5 mM), aldose reductase catalyzed reaction represents less than 3% of total glucose utilization [85]. However, hyperglycemia results in saturation of hexokinase and more than 30% of glucose is directed into the polyol pathway [86]. In a diabetic state, polyol pathway increases in tissues that do not require insulin for cellular glucose uptake, such as retina, kidney, peripheral nerves and blood vessels [87].

The overall reaction of the polyol pathway leads to a shortage of intracellular NAD(P)H and a surplus of NADH, i.e, a reductive imbalance. Increased NADH generation during conversion of sorbitol to fructose provides substrate for NADH oxidase to generate ROS [88]. NADH serves as a source of electrons in complex 1 of the electron transport chain resulting in increased mitochondrial generation of superoxide radical. In diabetic cells, oxidative phosphorylation in mitochondria is enhanced due to increase flux of electron donors into the electron transport chain. This drives the inner mitochondrial membrane potential upward causing blockage of electron transfer inside complex III [89]. Electrons back up to coenzyme Q results and electrons are transferred one at a time to molecular oxygen, generating superoxide. DNA damage by superoxide and peroxynitrite results in the activation of poly (ADP-ribose) polymerase (PARP), a DNA repair enzyme. PARP reduces the activity of glyceraldehyde-3- phosphate dehydrogenase (GAPDH) (an enzyme of the glycolytic pathway which catalyses the conversion of glyceraldehydes -3 phosphate to 1, 3 biphosphoglycerate) by ADP- ribosylation [90, 91]. A consequence of GAPDH inhibition by PARP is an increase in triose phosphate pool, upstream of GAPDH and increase flux of intermediates into the damaging pathways of diabetic complications.

The polyol pathway also results in reduction in the bioavailability of NAD(P)H. The reduced bioavailability of NAD(P)H negatively affects the antioxidant defence system by depleting glutathione (GSH) a very important antioxidant. This is because the activity of GSH reductase, an antioxidant enzyme that generates GSH from its oxidized form (GSSH) depends on NAD(P)H. Depletion of NAD(P)H also decreases the synthesis of nitric oxide (NO), a vaculoprotective agent. NAD(P)H serves as a cofactor for nitric oxide synthase (NOS) which synthesizes NO from L-arginine. If endothelial nitric oxide synthase (eNOS) lack its substrate, L-arginine or one of its co-factor, it may produce superoxide radical (.O2 -) instead of NO and this is referred to as ‘‘uncoupled state of nitric oxide’’[92]. Nitric oxide performs several physiological roles such as inhibition of platelet activation, vascular relaxation [93] and acts as an anti-inflammatory agent by reducing platelet aggregation and adhesion [94]. These properties inhibit atherogenesis and protect the blood vessel. Reduced bioavailability of NO level will therefore increase inflammation, enhance thrombosis and disrupt the integrity of endothelial cells. Reduction in NO has been documented in diabetes subjects with nephropathy [95]. Superoxide anion directly quenches NO by forming highly reactive peroxynitrite (ONOO-) which initiates lipid peroxidation, oxidizes sulfhydryl group in protein and nitrates amino acids such as tyrosine, thereby affecting many signal transduction pathways. The polyol pathway serves as a main source of ROS generation in the retina [96]. In addition, sorbitol accumulation has been implicated in osmotic swelling of the eye lens and cataractogenesis [97].

4.2. Advanced glycation end product (AGEs) formation and ROS generation in diabetic complications

Glucose can react spontaneously with free amino groups of protein to form Schiff bases. These Schiff bases through complex reactions such as amadori rearrangement, dehydration and condensation forms cross-linked heterogeneous fluorescent derivatives called advanced glycation end products (AGEs). Advanced glycation end products constitute a heterogeneous group of molecules formed by non-enzymatic reactions of reducing sugars, ascorbate and other carbohydrates with amino acids, lipids and nucleic acids [98, 99]. Glycation end product’s adducts such as pyraline, pentosidine and N- Carboxy- methyl lysine (CML) are found to be elevated in diabetic tissues [100 - 102].

Once formed, AGEs can cause tissue damage by two main pathways which are: (1) formation of cross links that alter protein structure and function and, (2) interaction of AGE with AGE-cell surface receptors on the surfaces of various cells such as endothelial cells, macrophages, neurons, and smooth-muscle cells resulting in activation of cell signaling and gene expression that induces oxidative stress and inflammation [98, 99; 102-105]. Oxidative stress can accelerate AGE formation while AGE formation can also amplify the production of more ROS resulting in a vicious cycle of AGE formation and oxidative stress.

AGE’s mediate some of their effect via interaction with some receptors that have been shown to bind to these chemical moieties. Among these receptors, Receptor for Advanced Glycation End products (RAGE) is the most extensively studied [106]. Evidence from numerous studies suggest that AGE’s are involved in a vicious cycle of inflammation, generation of ROS and increased production of AGE’s. Ligand RAGE interaction results in activation of pathways such as p21ras, erk1/2 (p44/p42), MAP kinases, p38 and SAPK/JNK MAP kinases [107-109]. A consequence of the activation of these pathways is the nuclear translocation of transcription factor, Nuclear Factor Kappa B (NF-КB). Translocation of NF-КB to the nucleus increases the transcription of a number of proteins such as, vascular endothelial growth factor (VEGF), monocyte chemoattractant protein-1 (MCP-1), vascular cell adhesion molecule-1 (VCAM-1) and intracellular adhesion molecule-1 (ICAM-1) and pro-inflammatory cytokines such as interleukin (IL)-1β, IL-6, 1L-18 and tumour necrosis factor (TNF)-α which are centrally involved in the endothelial recruitment of neutrophil and subsequent development or progression of atherosclerotic plaque [109-112].

The gene regions of NF-КB are located at the promoter region of RAGE. Moreover, binding of NF-КB to the promoter region of RAGE results in up-regulation of RAGE itsel. Interaction of AGE with RAGE generates more oxidative stress and this further potentiates the formation of AGE’s [109, 113]. Generation of ROS by ligand stimulated RAGE activation is mediated at least in part via activation of NADPH oxidase [114]. Other mechanisms by which AGE’s may be linked to increased generation of ROS is by reducing the activities of enzymatic antioxidant such as SOD and CAT, lowering of glutathione stores, and activation of PKC [107, 115, 116].

Increased renal AGE in diabetic animals and patients have been linked to structural abnormality observed in diabetic nephropathy such as mesangial expansion, glomerular basement membrane thickening and tubulointerstitial fibrosis [117]. Advanced Glycation End Product’s level is increased with decreased renal function in type 1 diabetic patients [118]. Evidence from clinical studies indicates a correlation between progression of diabetic retinopathy and the level of AGE in serum and retinal blood vessels of diabetic patients [100, 119]. In diabetes, increased AGE’s are observed within retinal capillary cells and causes pericyte loss in diabetic retinopathy [120]. AGE’s induce toxic effects on retinal pericytes by causing oxidative stress and subsequent apoptosis [121].

High levels of serum AGE’s have been documented in patients with type 2 diabetes mellitus and coronary heart disease [122]. Glycation increases susceptibility of low density lipoprotein (LDL) to oxidative modification which is considered a critical step in its atherogenicity [123]. Glycation end products can also enhance atherosclerosis by trapping LDL in the subendothelium and decrease the recognition of AGE-modified LDL by LDL receptor [124]. Modification of LDL and its increased localization in vessels increases foam cell production and accelerates atherosclerosis development [125]. Oxidative stress induces AGE’s formation on collagen leading to cross-linking which is considered to play a role in diabetic cardiomyopathy [126]. The intermolecular collagen cross-linking caused by AGE increases vascular stiffness and interferes with arterial blood flow [127, 128] and this partly explains the diastolic dysfunction and systolic hypertension seen in diabetic subjects.

4.3. Protein kinase C (PKC) activation and ROS generation in diabetic complications

PKC activation is related to vasoconstriction, proliferation and overgrowth of smooth muscle cells as well as accelerated synthesis of extracellular matrix proteins, and thus plays significant roles in the onset and progression of vascular cell dysfunction in diabetes mellitus [129-131]. Two major pathways have been implicated in the activation of PKC in hyperglycemia. Persistent and excessive activation of several PKC isoforms result primarily from enhanced de novo synthesis of diacylglycerol (DAG) from glucose via increase in triose phosphate availability [90, 105, 132, 133]. There is also evidence that the interaction between AGE’s and their cell-surface receptors can result in enhanced activity of PKC isoforms [134, 135].

PKC likely regulates diabetic complications on multiple levels such as activation of eNOS, NAD(P)H oxidase, phospholipase A2 (PLA2), endothelin-1 (ET-1), Vascular endothelial growth factor (VEGF), Transforming growth factor-β (TGF-β), and by activating NF-KB. Diacylglycerol activated PKC alters the gene expression of key proteins leading to decrease blood flow, capillary occlusion, inflammation, free radicals generation and damage to cellular macromolecule [130-132, 136, 137].

High glucose levels can stimulate ROS production via a PKC-dependent activation of NAD(P)H oxidase in cultured aortic endothelial cells, smooth muscle cells, and renal mesangial cells [84]. Nicotinamide adenine dinucleotide phosphate oxidase, which is primarily found in phagocytic cells, is the main source of ROS in non-phagocytic cells such as mesangial cells, endothelial cells [138], fibroblasts [139], podocytes [140] and smooth muscle cells [141]. The expression of NAD(P)H oxidase components is up-regulated in vascular tissues from animal models of diabetes and in patients with diabetes and coronary artery disease [142-144]. Experimental evidence indicates that NAD(P)H oxidase-dependent production of ROS may cause DNA damage in diabetic renal tissues leading to the development of nephropathy [145]. Increased activity of the NAD(P)H oxidase has also been reported in the retina of diabetic rats suggesting its involvement in the development of diabetic retinopathy [146].

Advertisement

5. Antioxidant as therapeutic agents in the management of diabetes mellitus

Despite efforts to control blood glucose, tissue and organ damage are cumulative over many years in most diabetic patients. Varying degrees of hyperglycemia are virtually unavoidable in subjects with diabetes mellitus and glycemic memory has been used to describe the development of diabetes-related complications in diabetic patients even after normoglycemia has been restored and initial glycemic environment is remembered in the target organs [105,147]. It is noteworthy that ROS has been implicated as a major cause of the metabolic memory after glucose normalization due to the chains of reactions leading to cell damage and loss of cellular function. Due to the implication of hyperglycemia-induced oxidative stress in diabetes, these patients should in theory benefit from antioxidant supplementation. The beneficial effect of antioxidants has been reported in animal models of diabetes and in diabetic patients [50, 148]

Advertisement

6. Vitamins

Vitamin E is a fat-soluble vitamin. It has been shown that plasma α-tocopherol concentrations are lower in diabetics compared to controls [58] and appear to be even lower in diabetics with complications such as microangiopathy than in diabetics without complications [81]. Administration of Vitamin E has proven to be beneficial in preventing cellular damage by inhibition of lipid peroxidation, protein oxidation, protein glycations and platelet aggregation [149-151] Vitamin E supplementation for two weeks (600 mg/day) lowered urinary F2-isoprostanes (a marker of lipid oxidation) in type 2 diabetics [152]. It was shown in a study that a decrease in plasma F2-isoprostanes was seen in type 2 diabetic patients after six weeks supplementation with Vitamin E [153].

Oxidative stress in the kidney of diabetics is usually associated with tissue damage that interferes with proper organ function, causing an increase in urinary protein excretion and blood urea nitrogen (BUN) [154]. Vitamin E supplementation (1000 IU/kg diet) to diabetic rats for 4 weeks significantly reduced urinary protein excretion and BUN suggesting a beneficial effect on kidney function [154]. Inhibitory effect of Vitamin E on glycation of hemoglobin in type I and type 2 diabetic rats has been documented [151, 155]. The ability of vitamin E to inhibit AGE’s might be due to its antioxidant effect on the autoxidative pathways of AGE formation [156]. Vitamin E administration has also reduced oxidation of low density lipoprotein (LDL) and development of atherosclerosis [157].

Numerous studies have shown that vitamin E normalized parameters of oxidative stress and inhibited vascular abnormalities caused by hyperglycemia-induced production of DAG and PKC activation in the retina, glomerulus and macrophages [158-160]. Supplementation with vitamin E reduced basement membrane thickening in diabetic rat retina and reduced vascular endothelial growth factor (VEGF) and aldose reductase activity, the abnormalities associated with diabetic retinopathy [161]. Dietary supplementation of vitamin E (2000 IU/kg) to diabetic rats for 8 weeks had cardioprotective effects which was simultaneously associated with an ability of vitamin E to blunt diabetes-induced amplification of myocardial 8-iso PGF2 and oxidized GSSG formation [162]. Clinical trials with vitamin E provided evidence that vitamin E may improve cardiovascular function [163, 164]. However, most large studies with vitamin E have not yielded positive benefits for decreasing the development or progression of diabetic microvascular and cardiovascular pathologies or mortality [165, 166].

Vitamin C is an antioxidant vitamin which plays an important role in protecting free radical-induced damage and a decrease in basal vitamin C levels has been documented in type 2 DM. Treatment of diabetic rats with vitamin C significantly decreased renal malondialdehyde, albuminuria, proteinuria, glomerular and tubulointerstitial sclerosis, suggesting the role of vitamin C in suppressing the progression of renal injury in diabetic rats [167]. Vitamin C also improved diabetes-induced endothelial dysfunction in a rat model by enhancing NO bioavailability [168].

The beneficial effects of vitamin C supplementation in humans are controversial. A study reported that vitamin C may improve glycemic control, lowering both fasting blood glucose and glycated haemoglobin (HbA1c) [169]. Chronic oral administration of vitamin C to patients with type 2 diabetes causes a decline in plasma free radicals that is associated with improved whole body glucose disposal [170,171] and improved endothelial function [172]. Recently, another study reported a reduction in the malondialdehyde (MDA) level, a major product of oxidative damage in both fasting and postprandial states of type 2 diabetic patients after vitamin C (1000 mg day-1) supplementation for 6 weeks although no effect was observed on lipid profiles [173]. Some studies have indicated that the intra-arterial infusion of vitamin c restores endothelium-dependent vasodilation in patients with type 1 or type 2 diabetes [174, 175] suggesting that hyperglycemia-induced oxidative stress mediates endothelial dysfunction in diabetic patients.

However, in contrast to these promising results, other studies showed no beneficial effect with vitamin C treatment. Chen and colleagues [176] concluded that a high oral dose of vitamin C therapy was ineffective at improving endothelial dysfunction and insulin resistance in type 2 DM. It is important to note that complete replenishment of vitamin C levels was not achieved in the subjects. This is crucial since high concentrations of vitamin C (>80 μM) has been documented as a requirement for the preservation of NO-dependent endothelial function as vitamin C only competes with NO for superoxide anion at these high concentrations [177-178]. Also, in another study, no beneficial effects of oral vitamin C supplementation (1.5 g daily for 3 weeks) was observed on blood pressure, oxidative stress, and endothelial function in type 2 diabetes [179].

Advertisement

7. Flavonoids

Flavonoids (bioflavonoids) are a diverse group of polyphenols (phenyl benzopyrans) which function as phytochemicals [180]. Flavonoids are well-known for their multi-directional biological activities including anti-diabetic efficacy. Experimental evidence has shown that flavonoids exhibits anti-inflammatory [181], anticarcinogenic [182], antiviral [183] and antiallergic properties. These effects are generally associated with free radical scavenging activity of flavonoids. The antioxidant effects of flavonoids are enhanced by the number and position of hydroxyl groups in the molecule. The catechol structure, presence of unsaturation and 4-oxo function in the C-ring also contributes to their radical scavenging activity [184, 185]. Flavonoids may be capable of binding the transition metal ions, which play a role in glycoxidation, thus preventing metal-catalysed formation of hydroxyl radicals or related species from H2O2 [186].

Figure 1.

Classes of flavonoids [187]

The potential beneficial effects of flavonoids in the prevention of diabetes mellitus and its associated complications have been investigated both in vitro and in vivo studies (Table 3). The inhibitory effect of flavonoids on glycation has been demonstrated and it is suggested that this effect is partly due to their antioxidant properties [188]. Epigallocatechin (EGC) has a beneficial effect in a rat model of diabetic nephropathy via suppressing hyperglycemia, proteinuria and lipid peroxidation. EGC also reduced renal accumulation of AGE’s and their related oxidative stress [189]. Another study demonstrated the in vitro inhibitory effect of different flavonoids on pentosidine formation in collagen in the presence of glucose (250 mmol/L). The decreasing inhibitiory activity was observed from myricetin, quercetin, rutin, catechin and kaempferol in a structure and concentration dependent manner [190]. Kim and colleagues [191] also investigated the effect of quercetin, isoquercitrin, hyperin and cacticin on formation of AGE’s in vitro. At a concentration of 50 mΜ, the percentages of inhibition were 1.0, 89.6, 92.0 and 40.5, respectively. The inhibitory effect of hyperin on AGE formation was 6.5 times higher than that of aminoguanidine, a known AGE inhibitor which showed 14.1% inhibition at a concentration of 50 μM.

Flavonoids, in addition to their antioxidant effect, possess inhibitory activity on aldose reductase pathway and can serve as a potential multifunctional agent in the prevention of diabetic retinopathy. Goodarzi et al. [192] showed that oral administration of quercetin and the flavanone, naringin to streptozotocin-induced diabetic rats significantly reduced aldose reductase activity in the lenses compared to control. Oral administration of two isoflavone compounds, tectorigenin and irigenin also inhibited sorbitol accumulation in the lenses of streptozotocin induced diabetic rats [193].

Activation of PKC contributes to the loss of capillary pericytes and thickening of vascular basement membrane (BM) in diabetic retinopathy [194]. Also PKC mediated alterations in vascular permeability, blood flow, formation and response to angiogenic growth factors contribute to retinal leakage, ischemia, and neovascularisation [195]. Therefore, PKC inhibitors can be targeted for the treatment of diabetic retinopathy. Hesperetin (Hsp), a flavanone found in citrus fruits and a potent antioxidant has retina vasculo-protective properties due to its strong anti-angiogenic effect via inhibiting VEGF and PKC-β pathways [196]. Modulation of endogenous biomarkers and inhibition of diabetes induced neuropathic pain was observed in diabetic rats after naringin (4′,5,7-trihydroxy flavonone 7-rhamnoglucoside) administration [197]. In the same study, a dose dependent decrease in the levels of oxidative-nitrosative stress, inflammatory mediators as well as apoptosis was documented in neural cells. The antioxidant properties of naringin may be a factor in the inhibition of neurodegeneration.

The soy isoflavone genistein (3 and 6mg/kg), administered by a subcutaneous injection to diabetic mice relieved peripheral painful neuropathy by reverting the proinflammatory cytokine and ROS overproduction. It also restored the inducible nitric oxide synthase (iNOS) and eNOS content and increased NO production in thoracic aorta although treatment had no effect on hyperglycemia [198]. The flavonoid luteolin (200 mg/kg), when administered to rats orally, protected against the progression of diabetes-induced cardiac dysfunction by attenuation of myocardial oxidative stress probably through its antioxidant properties [199].

In a double blind placebo-controlled study, the effects of daflon 500 (made up of flavonoids diosmin (90%) and hesperidin (10%)) was investigated in a group of 28 type 1 diabetic patients. Treatment with these flavonoids resulted in a decrease in HbA1C which is associated with an increase in the level and activities of thiol-containing antioxidants such as glutathione peroxidase [200]. The in vitro protective effect of myricetin on protein oxidation and membrane lipid peroxidation of erythrocytes from diabetic patients was reported in a study by Pandey and co-workers [201].

The treatment of diabetic rats with rutin decreased fasting plasma glucose, glycosylated haemoglobin, thiobarbituric acid reactive substances and lipid hydroperoxides while levels of non-enzymatic antioxidants were increased [202]. In another study, rutin supplementation (500 mg tablets) to diabetic patients for 60 days decreased the levels of fasting blood glucose, blood pressure and improved lipid profiles in the diabetic subjects [203]. Rutin reduced blood glucose, ameliorated oxidative stress and inhibited the accumulation of extracellular matrix (ECM) component and glomerular basement membrane thickening in the renal cortex of diabetic rats suggesting its renoprotective effect in experimental diabetic nephropathy [204]. The inhibitory effect of rutin on AGE formation in STZ-induced rats has also been shown [205].

Diosmin (DS) (diosmetin 7-O-rutinoside) is a natural flavone glycoside which can be obtained by dehydrogenation of the corresponding flavanone glycoside, hesperidin that is abundant in the pericarp of various citrus fruits [206]. Diosmin treatment of streptozotocin-nicotinamide induced diabetic rats, ameliorated oxidative stress in plasma and tissues as evidenced by improved glycemic and antioxidant status along with decreased lipid peroxidation [207]. Experimental evidence showed the potential of rutin, a flavonol to delay glomerulosclerosis of diabetic nephropathy (DN) due to its ability to inhibit cell hypertrophy and the accumulation of ECM mediated by TGF-β1/Smads and ROS signals in mesangial cells cultured by high glucose [208] Quercetin enhances endothelium-derived NO bioavailability, reduced blood glucose levels and oxidative stress in diabetic rats suggesting its beneficial effect in vascular function [209].

Classes of Flavonoid and Food sources Selected examples Target organ, tissue or cells Mechanism of action Reference
Flavonols
(Brussel sprouts, apples, onion, curly kale, leek, beans, cherries, Citrus fruits, Cranberries)
Morin Liver Decreased MDA levels ,
Increased activity of SOD and GSH concentration.
[210]
Hepatocytes Decreased ROS production, DNA damage and apoptosis. Modulation of antioxidant enzymes; GSH, CAT, SOD and GPX. [211]
Rutin Kidney, serum,
urine
Lowered blood glucose and improved renal function. Increased total antioxidant capability activities of SOD, CAT and GPX. Lowered ECM accumulation and AGE formation. Decreased renal expression of TGF-β. [204]
Quercetin Pancreas, serum, erythrocyte. Lowered MDA and NO level, increased antioxidant enzyme activity and preservation of islet cells integrity. [212]
Kidney , serum, urine Lowered blood glucose and improved renal function. Reduced renal lipid peroxides and increased activity of anti-oxidative enzymes; SOD and CAT and non-enzymatic antioxidant GSH. [213]
Flavanones (Citus peel, Orange juice, grape fruit juice,
lemon juice)
Hesperidin & Naringin Liver, serum Boost antioxidant system by increasing activities of SOD, GR, GPx, CAT and levels of non-enzymatic antioxidants; GSH, VIT-C and VIT-E.
Decreased lipid peroxidation product, MDA and proinflammatory markers, TNF-α, IL-6.
[214]
Hesperidin Retina, plasma Decreased aldose reductase activity and levels of AGE’s, VEGF, ICAM-1, TNF-α, IL-1β and MDA while increasing SOD activity. [215]
Naringenin Pancreas, heamoglobin, serum, plasma Lowered fasting blood glucose, decreased hyperglycemia, glycated haemoglobin, MDA and markers of hepatic damage.
Increased levels of insulin and enzymatic and non-enzymatic antioxidants.
[216]
Kidney, liver serum, urine, Improved glycemic control and elevated insulin level, reduced plasma levels of kidney dysfunction markers. Lowered renal activity and expression of NF-KB and pro-inflammatory cytokine and chemokine, suppression of PKC activity. [217]
Flavanolols
(Milk thistle, red onion,
Siberian larch tree)
Silymarin Kidney Increased expression and activity of SOD, GPX, CAT. Decreased high blood glucose [218]
Flavones
(Parsley, pepper celery, broccoli capsicum)
Luteolin Kidney Decreased activity of SOD, MDA content and expression of Heme Oxygenase-1 (HO-1) protein. [219]
Aortic ring Aortic Vasorelaxation, decrease in ROS production, increased activity of SOD, NOS and level of NO. [220]
Diosmin Liver and kidney Decreased TBARS and hydroperoxides.
Increased activity of enzymatic antioxidants ;SOD, CAT, GPx, GST and GR and non-enzymatic antioxidants; GSH, Vitamin C and Vitamin E.
[207]
Flavones Chrysin and luteolin Serum and aorta Aortic relaxation, decreased blood pressure, decreased lipidemia and serum AGE’s.
Increased .NO generation
[221]
Apigenin Serum and liver Increased insulin levels and decreased hyperglycemia. Normalized LPO and endogeneous antioxidants, CAT, SOD, GSH in the liver. [222]
Flavan-3-ols (Red wine and red grapes, green and black tea) Catechin Thoracic aorta Decreased hyperglycemia NADPH oxidase activity and ROS production. Increased insulin level, lowered blood pressure and improved aortic relaxation. [223]
Epicatechin Pancreatic Islets, plasma, haemoglobin Increased anti-inflammatory cytokines, IL-10, IL-12.
Improved glucose tolerance and insulin levels and lowered HbA1C.
[224]
Isoflavones
(Soy foods and legumes)
Genistein Kidney Decreased MDA level and expression of PKC and pro-inflammatory proteins such as NF-KB, MCP-1 and Cox-2.
Activation of antioxidant enzymes and defense against oxidative damage via increase expression of Nrf2, a transactivator of antioxidant genes.
[225]
Daidzein Aorta Maintenance of endothelium dependent relaxation and attenuation of oxidative stress via decrease MDA levels and increase SOD activity [226]

Table 3.

Beneficial effects of some flavonoids in diabetes mellitus

Abbreviations: Find all citations in this journal (defECM: Extracellular matrix, MDA: Malondialdehyde, NO: Nitric oxide, NOS: Nitric oxide synthase, NADPH; nicotinamide adenine dinucleotide phosphate, LPO: Lipid peroxidation, Nrf2: NF-E2-related factor-2, Cox-2: Cyclooxygenase-2, TAOC: Total antioxidative capability


The inherent antioxidative properties of some common antidiabetic drugs such as aminoguanidine, statins, thiazolidinediones, glibenclamide and repaglinide also provides an additional support to the involvement of oxidative stress in diabetes and therefore suggest that the use of antioxidants as therapeutic agents in diabetes is a promising approach [227-231].

Advertisement

8. Conclusion and future perspective

Increased ROS production has been suggested as a common pathway linking diverse pathogenic mechanisms of diabetic vascular complications. There are numerous evidences from animal studies on the beneficial effect of antioxidant vitamins supplementation in diabetes mellitus, but results from clinical studies are inconclusive. The antioxidant activity of some anti-diabetic drugs has also been shown to contribute significantly to their therapeutic effect. Biflavonoids as antioxidants are promising and attractive natural substances to enrich the current therapy options against diabetes. The overall positive results from animal studies suggest that the role of antioxidants cannot be underestimated in the quest to find effective therapies for diabetic complications. A multi-therapeutic approach to the treatment of diabetic complications might increase the chance of successful therapeutic intervention. In addition to maintaining glycemic control, blockage of pathways involved in the formation of free radicals with antioxidants is a promising approach to the treatment of hyperglycemia-mediated complications in humans. The bioactivity of flavonoids in vivo can be greatly influenced by metabolism and bioavailability and this information is limited in many studies. Limited clinical studies have been carried out to support the beneficial effects of flavonoids in the prevention of diabetic complications. In order to achieve the goal of using flavonoids in the management of diabetes mellitus, further investigation on the long and short-term effect of the ingestion of flavonoids in humans is warranted.

References

  1. 1. Bastaki, A. 2005. Diabetes mellitus and its treatment. Int J Diab and Metab 13(3): 111.
  2. 2. Zhao, Y. 2011. Autoimmunity and Therapeutic Challenges of Type 1 Diabetes. Transl Med,1: 104e
  3. 3. Ruhe, R.C. & McDonald, R.B. 2001. Use of antioxidant nutrients in the prevention and treatment of type 2 diabetes. J Am Coll Nutr, 20 (sup5): 363S-369S.
  4. 4. Resnick, H.E. & Howard, B.V. 2002. Diabetes and cardiovascular disease. Annu Rev Med, 53(1): 245-267.
  5. 5. Fowler, M.J. 2008. Microvascular and macrovascular complications of diabetes. Clin Diabetes, 26(2): 77-82.
  6. 6. Cook, M.N., Girman, C.J., Stein, P.P., Alexander, C.M. & Holman, R.R. 2005. Glycemic control continues to deteriorate after sulfonylureas are added to metformin among patients with type 2 diabetes. Diabetes Care, 28(5): 995-1000.
  7. 7. Kowluru, R.A., Abbas, S.N. & Odenbach, S. 2004. Reversal of hyperglycemia and diabetic nephropathy: effect of reinstitution of good metabolic control on oxidative stress in the kidney of diabetic rats. J Diabetes Complication, 18(5): 282-288.
  8. 8. Mayor, S. 2008. Intensive glucose lowering arm of diabetes trial is stopped after excess deaths. Br Med J, 336(7641): 407.
  9. 9. Ismail-Beigi, F., Craven, T., Banerji, M.A., Basile, J., Calles, J., Cohen, R.M., Cuddihy, R., Cushman, W.C., Genuth, S. & Grimm Jr, R.H. 2010. Effect of intensive treatment of hyperglycaemia on microvascular outcomes in type 2 diabetes: an analysis of the ACCORD randomised trial. The Lancet, 376(9739): 419-430.
  10. 10. Kawahito, S., Kitahata, H. & Oshita, S. 2009. Problems associated with glucose toxicity: Role of hyperglycemia-induced oxidative stress. World J Gastroenterol: 15(33): 4137.
  11. 11. Ceriello, A. 2003. New insights on oxidative stress and diabetic complications may lead to a “causal” antioxidant therapy. Diabetes Care, 26(5): 1589-1596.
  12. 12. Giacco, F. & Brownlee, M. 2010. Oxidative stress and diabetic complications. Circ Res, 107(9): 1058-1070.
  13. 13. Zhang, C., Liu, J., Pan, H., Yang, X. & Bian, K. 2011. Mitochondrial dysfunction induced by excessive ROS/RNS-metabolic cardiovascular disease and traditional Chinese medicines intervention. China J Chinese Mat med, 36(17): 2423.
  14. 14. Jain, M. 2012. Histopathological changes in diabetic kidney disease. Clin Queries: Nephrol, 1(2): 127-133.
  15. 15. Lewis, E.J. & Xu, X. 2008. Abnormal Glomerular Permeability Characteristics in Diabetic Nephropathy Implications for the therapeutic use of low–molecular weight heparin. Diabetes Care, 31(Supplement 2): S202-S207.
  16. 16. O’Connor, A.S. & Schelling, J.R. 2005. Diabetes and the kidney. Am J Kidney Dis, 46(4): 766-773.
  17. 17. Marshall, S.M.m& Flyvbjerg, A. 2006. Prevention and early detection of vascular complications of diabetes. Br Med J, 333(7566): 475.
  18. 18. Santos, J.M., Mohammad, G., Zhong, Q. & Kowluru, R.A. 2011. Diabetic retinopathy, superoxide damage and antioxidants. Curr Pharm Biotechnol 1;12(3):352-61
  19. 19. Engerman, R.L. 1989. Pathogenesis of diabetic retinopathy. Diabetes, 38 (10): 1203-1206.
  20. 20. Hammes, H. 2005. Pericytes and the pathogenesis of diabetic retinopathy. Horm Metab Res, 37(S 1): 39-43.
  21. 21. American Diabetes Association: Standards of medical care in diabetes 2007 [Position Statement]. Diabetes Care 30:S4–S41, 2007
  22. 22. Pazdro, R. & Burgess, J.R. 2010. The role of vitamin E and oxidative stress in diabetes complications. Mech Ageing Dev, 131(4): 276-286.
  23. 23. Obrosova, I.G. 2009. Diabetes and the peripheral nerve. Biochim Biophys Acta, 1792(10): 931-940.
  24. 24. Soumya, D. 2011. Late Stage Complications of Diabetes and Insulin Resistance. J Diabetes Metabol, 2:167
  25. 25. Schwartz, C.J., Valente, A.J., Sprague, E.A., Kelley, J.L., Cayatte, A.J. & Rozek, M.M. 1992. Pathogenesis of the atherosclerotic lesion: implications for diabetes mellitus. Diabetes Care, 15(9): 1156-1167.
  26. 26. American Diabetes association. Consensus Statement, 1993. Role of cardiovascular risk factors in prevention and treatment of macrovascular disease in diabetes. Diabetes Care. 16:72-78.
  27. 27. Orchard, T.J., Costacou, T., Kretowski, A. & Nesto, R.W. 2006. Type 1 diabetes and coronary artery disease. Diabetes Care, 29(11): 2528-2538.
  28. 28. Ding, H. & Triggle, C.R. 2005. Endothelial cell dysfunction and the vascular complications associated with type 2 diabetes: assessing the health of the endothelium. Vasc Health Risk Manag, 1(1): 55.
  29. 29. Winer, N. & Sowers, J.R. 2004. Epidemiology of diabetes. J Clin Pharmacol, 44(4): 397-405.
  30. 30. Epstein, F.H. & Ross, R. 1999. Atherosclerosis-an inflammatory disease. New Engl J Med, 340(2): 115-126.
  31. 31. Libby, P., Ridker, P.M. & Hansson, G.K. 2011. Progress and challenges in translating the biology of atherosclerosis. Nature, 473(7347): 317-325.
  32. 32. Funk, S.D., Yurdagul, A. & Orr, A.W. 2012. Hyperglycemia and endothelial dysfunction in atherosclerosis: lessons from type 1 diabetes. Int J Vasc Med, 2012:1-19.
  33. 33. Creager, M.A., Lüscher, T.F., Cosentino, F. & Beckman, J.A. 2003. Diabetes and vascular disease pathophysiology, clinical consequences, and medical therapy: part I. Circulation, 108(12): 1527-1532.
  34. 34. Valko, M., Leibfritz, D., Moncol, J., Cronin, M.T., Mazur, M. & Telser, J. 2007. Free radicals and antioxidants in normal physiological functions and human disease. Int J Biochem Cell Biol, 39(1): 44-84.
  35. 35. Dröge, W. 2002. Free radicals in the physiological control of cell function. Physiol Rev, 82(1): 47-95.
  36. 36. Pacher, P., Beckman, J.S. & Liaudet, L. 2007. Nitric oxide and peroxynitrite in health and disease. Physiol Rev, 87(1): 315-424.
  37. 37. Sachdev, S. & Davies, K.J. 2008. Production, detection, and adaptive responses to free radicals in exercise. Free Radic Biol Med, 44(2): 215-223.
  38. 38. Sies, H. 1997. Oxidative stress: Oxidants and antioxidants. Exp Physiol., 82, 291–295.
  39. 39. Halliwell, B. & Gutteridge, J. M. C., 2007. Free Radic Biol Med. 4th. Edn, Clarendon Press, Oxford
  40. 40. Bahorun, T., Soobrattee, M., Luximon-Ramma, V. & Aruoma, O. 2006. Free radicals and antioxidants in cardiovascular health and disease. Internet J Med Update, 1(2): 25-41.
  41. 41. Halliwell, B. 2007. Biochemistry of oxidative stress. Biochem Soc Trans, 35(Pt 5): 1147-1150.
  42. 42. Abdollahi, M., Ranjbar, A., Shadnia, S., Nikfar, S. & Rezaie, A. 2004. Pesticides and oxidative stress: a review. Medical science monitor: Int Med J Exp and Clin Res, 10(6): RA141-7.
  43. 43. Ridnour, L.A., Thomas, D.D., Mancardi, D., Espey, M.G., Miranda, K.M., Paolocci, N., Feelisch, M., Fukuto, J. & Wink, D.A. 2004. The chemistry of nitrosative stress induced by nitric oxide and reactive nitrogen oxide species. Putting perspective on stressful biological situations. Biol Chem, 385(1): 1-10.
  44. 44. Halliwell, B. 2011. Free radicals and antioxidants. Trends Pharmacol Sci, 32(3): 125-130.
  45. 45. Fukai, T. & Ushio-Fukai, M. 2011. Superoxide dismutases: role in redox signaling, vascular function, and diseases. Antioxid Redox Signal, 15(6): 1583-1606.
  46. 46. Lubos, E., Loscalzo, J. & Handy, D.E. 2011. Glutathione peroxidase-1 in health and disease: from molecular mechanisms to therapeutic opportunities. Antioxid Redox Signal, 15(7): 1957-1997.
  47. 47. Pham-Huy, L.A., He, H. & Pham-Huy, C. 2008. Free radicals, antioxidants in disease and health. Int J Biomed Sc: 4(2): 89.
  48. 48. Winterbourn, C.C. 1993. Superoxide as an intracellular radical sink. Free Radic Biol Med, 14(1): 85-90.
  49. 49. Jurkovič, S., Osredkar, J. & Marc, J. 2008. Molecular impact of glutathione peroxidases in antioxidant processes. Biochem Med, 18(2): 162-174.
  50. 50. Maritim, A., Sanders, R. & Watkins, 3.J. 2003. Diabetes, oxidative stress, and antioxidants: a review. J Biochem and Mol Tox, 17(1): 24-38.
  51. 51. Zelko, I.N., Mariani, T.J. & Folz, R.J. 2002. Superoxide dismutase multigene family: a comparison of the CuZn-SOD (SOD1), Mn-SOD (SOD2), and EC-SOD (SOD3) gene structures, evolution, and expression. Free Radic Biol Med, 33(3): 337-349.
  52. 52. Livingstone, C. & Davis, J. 2007. Review: Targeting therapeutics against glutathione depletion in diabetes and its complications. Br J Diabetes Vasc Dis, 7(6): 258-265.
  53. 53. May, J.M., Qu, Z. & Morrow, J.D. 1996. Interaction of Ascorbate and-Tocopherol in Resealed Human Erythrocyte Ghosts. Transmembrane electron transfer and protection from lipid peroxidation. J Biol Chem, 271(18): 10577-10582.
  54. 54. Smith, A., Shenvi, S., Widlansky, M., Suh, J. & Hagen, T. 2004. Lipoic acid as a potential therapy for chronic diseases associated with oxidative stress. Curr Med Chem, 11(9): 1135-1146.
  55. 55. Inoguchi, T., Li, P., Umeda, F., Yu, H.Y., Kakimoto, M., Imamura, M., Aoki, T., Etoh, T., Hashimoto, T. & Naruse, M. 2000. High glucose level and free fatty acid stimulate reactive oxygen species production through protein kinase C--dependent activation of NAD (P) H oxidase in cultured vascular cells. Diabetes, 49(11): 1939-1945.
  56. 56. Lee JL, Suh KS, Choi MC, Chon S, Oh S, Woo JT, Kim SW,Kim JW, Kim YS (2010). Kaempferol protects HIT-15 pancreatic beta-cells from 2-deoxy-D-ribose-induced oxidative damage. Phytother Res 24:419-423
  57. 57. Rösen, P., Nawroth, P., King, G., Möller, W., Tritschler, H. & Packer, L. 2001. The role of oxidative stress in the onset and progression of diabetes and its complications: asummary of a Congress Series sponsored byUNESCO‐MCBN, the American Diabetes Association and the German Diabetes Society. Diabetes Metabol Res Rev, 17(3): 189-212.
  58. 58. Nourooz-Zadeh, J., Rahimi, A., Tajaddini-Sarmadi, J., Tritschler, H., Rosen, P., Halliwell, B. & Betteridge, D. 1997. Relationships between plasma measures of oxidative stress and metabolic control in NIDDM. Diabetologia, 40(6): 647-653.
  59. 59. Brownlee, M. 2001. Biochemistry and molecular cell biology of diabetic complications. Nature, 414(6865): 813-820.
  60. 60. Evans, J.L., Goldfine, I.D., Maddux, B.A. & Grodsky, G.M. 2002. Oxidative stress and stress-activated signaling pathways: a unifying hypothesis of type 2 diabetes. Endocr Rev, 23(5): 599-622.
  61. 61. Shih, C., Wu, Y. & Lin, W. 2002. Antihyperglycaemic And Anti‐Oxidant Properties Of Anoectochilus Formosanus in Diabetic Rats. Clin Exp Pharmacol Physiol, 29(8): 684-688.
  62. 62. Manna, P., Das, J., Ghosh, J. & Sil, P.C. 2010. Contribution of type 1 diabetes to rat liver dysfunction and cellular damage via activation of NOS, PARP, IκBα/NF-κB, MAPKs, and mitochondria-dependent pathways: Prophylactic role of arjunolic acid. Free Radic Biol Med, 48(11): 1465-1484.
  63. 63. Hamden, K., Boujbiha, M.A., Masmoudi, H., Ayadi, F.M., Jamoussi, K. & Elfeki, A. 2009. Combined vitamins (C and E) and insulin improve oxidative stress and pancreatic and hepatic injury in alloxan diabetic rats. Biomed Pharmacother, 63(2): 95-99.
  64. 64. Prabakaran D, Ashokkumar N. Protective effect of esculetin on hyperglycemia-mediated oxidative damage in the hepatic and renal tissues of experimental diabetic rats. Biochimie. 2013 95(2):366-73.
  65. 65. Erejuwa, O.O., Sulaiman, S.A., Wahab, M.S.A., Salam, S.K.N., Salleh, M.S.M. & Gurtu, S. 2011. Comparison of antioxidant effects of honey, glibenclamide, metformin, and their combinations in the kidneys of streptozotocin-induced diabetic rats. Int J Mol Sci, 12(1): 829-843.
  66. 66. Aragno, M., Mastrocola, R., Medana, C., Restivo, F., Catalano, M.G., Pons, N., Danni, O. & Boccuzzi, G. 2005. Up-regulation of advanced glycated products receptors in the brain of diabetic rats is prevented by antioxidant treatment. Endocrinology, 146(12): 5561-5567.
  67. 67. Kowluru, R.A. & Kanwar, M. 2007. Effects of curcumin on retinal oxidative stress and inflammation in diabetes. Nutr and metabol, 4: 8
  68. 68. Babu, P.V.A., Sabitha, K.E. & Shyamaladevi, C.S. 2006. Therapeutic effect of green tea extract on oxidative stress in aorta and heart of streptozotocin diabetic rats. Chem Biol Interac, 162(2): 114-120.
  69. 69. Ramesh, B., Viswanathan, P. & Pugalendi, K.V. 2007. Protective effect of Umbelliferone on membranous fatty acid composition in streptozotocin-induced diabetic rats. Eur J Pharmacol, 566(1): 231-239.
  70. 70. Haidara, M.A., Mikhailidis, D.P., Rateb, M.A., Ahmed, Z.A., Yassin, H.Z., Ibrahim, I.M. & Rashed, L.A. 2009. Evaluation of the effect of oxidative stress and vitamin E supplementation on renal function in rats with streptozotocin-induced Type 1 diabetes. J Diabetes and Its Complications, 23(2): 130-136.
  71. 71. Rajavel, V., Sattar, A., Zubaid, M., Abdulla, M.A., Kassim, N.M. & Abdullah, N.A. 2012. Chronic Administration of Oil Palm (Elaeis guineensis) Leaves Extract Attenuates Hyperglycaemic-Induced Oxidative Stress and Improves Renal Histopathology and Function in Experimental Diabetes. Evid Base Compl Alternative Med, doi:10.1155/2012/195367
  72. 72. Park, K.S., Kim, J.H., Kim, M.S., Kim, J.M., Kim, S.K., Choi, J.Y., Chung, M.H., Han, B., Kim, S.Y. & Lee, H.K. 2001. Effects of insulin and antioxidant on plasma 8-hydroxyguanine and tissue 8-hydroxydeoxyguanosine in streptozotocin-induced diabetic rats. Diabetes, 50(12): 2837-2841.
  73. 73. Figarola, J., Scott, S., Loera, S., Tessler, C., Chu, P., Weiss, L. & Hardy, J. 2003. LR-90 a new advanced glycation endproduct inhibitor prevents progression of diabetic nephropathy in streptozotocin-diabetic rats. Diabetologia, 46(8): 1140-1152.
  74. 74. Likidlilid A, Patchanans N, Peerapatdit T, Sriratanasathavorn C, 2010. Lipid Peroxidation and Antioxidant Enzyme Activities in Erythrocytes of Type 2 Diabetic Patients. J Med Assoc Thai, 93 (6): 682-93
  75. 75. Sekhar, R.V., McKay, S.V., Patel, S.G., Guthikonda, A.P., Reddy, V.T., Balasubramanyam, A. & Jahoor, F. 2011. Glutathione synthesis is diminished in patients with uncontrolled diabetes and restored by dietary supplementation with cysteine and glycine. Diabetes Care, 34(1): 162-167.
  76. 76. Flores L, Rodela S, Abian J, Clària J, Esmatjes E, 2004 F2 isoprostane is already increased at the onset of type 1 diabetes mellitus: effect of glycemic control. Metabolism. 2004;53(9):1118-1120.
  77. 77. Selvaraj, N., Bobby, Z. & Sathiyapriya, V. 2006. Effect of lipid peroxides and antioxidants on glycation of hemoglobin: an in vitro study on human erythrocytes. Clin Chim Acta, 366(1): 190-195.
  78. 78. Goodarzi, M.T., Navidi, A.A., Rezaei, M. & Babahmadi‐Rezaei, H. 2010. Oxidative damage to DNA and lipids: correlation with protein glycation in patients with type 1 diabetes. J Clin Lab Anal, 24(2): 72-76.
  79. 79. Hinokio, Y., Suzuki, S., Hirai, M., Suzuki, C., Suzuki, M. & Toyota, T. 2002. Urinary excretion of 8-oxo-7, 8-dihydro-2′-deoxyguanosine as a predictor of the development of diabetic nephropathy. Diabetologia, 45(6): 877-882.
  80. 80. Ceriello, A., Mercuri, F., Quagliaro, L., Assaloni, R., Motz, E., Tonutti, L. & Taboga, C. 2001. Detection of nitrotyrosine in the diabetic plasma: evidence of oxidative stress. Diabetologia, 44(7): 834-838.
  81. 81. Martín-Gallán, P., Carrascosa, A., Gussinyé, M. & Domínguez, C. 2003. Biomarkers of diabetes-associated oxidative stress and antioxidant status in young diabetic patients with or without subclinical complications. Free Rad Biol Med, 34(12): 1563-1574.
  82. 82. Chung, S.S., Ho, E.C., Lam, K.S. & Chung, S.K. 2003. Contribution of polyol pathway to diabetes-induced oxidative stress. Journal of the American Society of Nephrology, 14(suppl 3): S233-S236.
  83. 83. Baynes, J.W. & Thorpe, S.R. 1999. Role of oxidative stress in diabetic complications: a new perspective on an old paradigm. Diabetes, 48(1): 1-9.
  84. 84. Inoguchi, T., Sonta, T., Tsubouchi, H., Etoh, T., Kakimoto, M., Sonoda, N., Sato, N., Sekiguchi, N., Kobayashi, K. & Sumimoto, H. 2003. Protein kinase C–dependent increase in reactive oxygen species (ROS) production in vascular tissues of diabetes: role of vascular NAD (P) H oxidase. Clin J Am Soc Nephrol, 14(suppl 3): S227-S232.
  85. 85. Morrison, A.D., Clements Jr, R.S., Travis, S.B., Oski, F. & Winegrad, A.I. 1970. Glucose utilization by the polyol pathway in human erythrocytes. Biochem Biophys Res Comm, 40(1): 199-205.
  86. 86. González, R.G., Barnett, P., Aguayo, J., Cheng, H. & Chylack, L. 1984. Direct measurement of polyol pathway activity in the ocular lens. Diabetes, 33(2): 196-199.
  87. 87. Stephen S.M. Chung, Eric C.M. HO, Karen S.L. Lam, and Sookja K. Chung, 2003. Contribution of Polyol Pathway to Diabetes-Induced Oxidative Stress. J Am Soc Nephrol 14: S233–S236
  88. 88. Morre, D.M., Lenaz, G. & Morre, D.J. 2000. Surface oxidase and oxidative stress propagation in aging. J Exp Biol, 203(10): 1513-1521.
  89. 89. Yang, X. & Trumpower, B.L. 1988. Protonmotive Q cycle pathway of electron transfer and energy transduction in the three-subunit ubiquinol-cytochrome c oxidoreductase complex of Paracoccus denitrificans. J Biol Chem, 263(24): 11962-11970.
  90. 90. Du, X., Matsumura, T., Edelstein, D., Rossetti, L., Zsengellér, Z., Szabó, C. & Brownlee, M. 2003. Inhibition of GAPDH activity by poly (ADP-ribose) polymerase activates three major pathways of hyperglycemic damage in endothelial cells. J Clin Investig, 112(7): 1049-1057.
  91. 91. Soriano, F.G., Virág, L., Jagtap, P., Szabó, É., Mabley, J.G., Liaudet, L., Marton, A., Hoyt, D.G., Murthy, K.G. & Salzman, A.L. 2001. Diabetic endothelial dysfunction: the role of poly (ADP-ribose) polymerase activation. Nature Med, 7(1): 108-113.
  92. 92. Vásquez-Vivar, J., Kalyanaraman, B., Martásek, P., Hogg, N., Masters, B.S.S., Karoui, H., Tordo, P. & Pritchard, K.A. 1998. Superoxide generation by endothelial nitric oxide synthase: the influence of cofactors. Proc Natl Acad Sci, 95(16): 9220-9225.
  93. 93. Mehta, J.L., Rasouli, N., Sinha, A.K. & Molavi, B. 2006. Oxidative stress in diabetes: a mechanistic overview of its effects on atherogenesis and myocardial dysfunction. Int J Biochem Cell Biol, 38(5): 794-803.
  94. 94. Sharma, J., Al-Omran, A. & Parvathy, S. 2007. Role of nitric oxide in inflammatory diseases. Inflammopharmacology, 15(6): 252-259.
  95. 95. Tessari, P., Cecchet, D., Cosma, A., Vettore, M., Coracina, A., Millioni, R., Iori, E., Puricelli, L., Avogaro, A. & Vedovato, M. 2010. Nitric oxide synthesis is reduced in subjects with type 2 diabetes and nephropathy. Diabetes, 59(9): 2152-2159.
  96. 96. Mara, L. & Oates, P.J. 2008. Diabetic Retinopathy. In The Polyol Pathway and Diabetic Retinopathy. : Springer: 159-186. Contemporary Diabetes 2008:159-186
  97. 97. Patel, D., Prasad, S., Kumar, R. & Hemalatha, S. 2011. Cataract: A major secondary complication of diabetes, its epidemiology and an overview on major medicinal plants screened for anticataract activity. Asian Pac J Tropical Dis, 1(4): 323-329.
  98. 98. Vlassara, H. & Palace, M. 2002. Diabetes and advanced glycation endproducts. J Intern Medicine, 251(2): 87-101.
  99. 99. Peppa, M., Uribarri, J. & Vlassara, H. 2004. The role of advanced glycation end products in the development of atherosclerosis. Curr Diabetes Rep, 4(1): 31-36.
  100. 100. Stitt, A.W. 2001. Advanced glycation: an important pathological event in diabetic and age related ocular disease. Br J Ophthalmol, 85(6): 746-753.
  101. 101. Wautier, J. & Guillausseau, P. 2001. Advanced glycation end products, their receptors and diabetic angiopathy. Diabetes Metabol, 27(5; PART 1): 535-544.
  102. 102. Ahmed, N., 2005. Advanced glycation end products-role in pathology of diabetic complications. Diabetes Res Clin Pract 67, 3-21.
  103. 103. Nishikawa, T., Edelstein, D., Du, X.L., Yamagishi, S., Matsumura, T., Kaneda, Y., Yorek, M.A., Beebe, D., Oates, P.J. & Hammes, H. 2000. Normalizing mitochondrial superoxide production blocks three pathways of hyperglycaemic damage. Nature, 404(6779): 787-790.
  104. 104. Sourris, K.C. & Forbes, J.M. 2009. Interactions between advanced glycation end-products (AGE) and their receptors in the development and progression of diabetic nephropathy-are these receptors valid therapeutic targets. Curr Drug Targets, 10(1): 42-50.
  105. 105. Giacco, F. & Brownlee, M. 2010. Oxidative stress and diabetic complications. Circ Res, 107(9): 1058-1070.
  106. 106. Ramasamy, R., Vannucci, S.J., Du Yan, S.S., Herold, K., Yan, S.F. & Schmidt, A.M. 2005. Advanced glycation end products and RAGE: a common thread in aging, diabetes, neurodegeneration, and inflammation. Glycobiology, 15(7): 16R-28R.
  107. 107. Yan, S.D., Schmidt, A.M., Anderson, G.M., Zhang, J., Brett, J., Zou, Y.S., Pinsky, D. & Stern, D. 1994. Enhanced cellular oxidant stress by the interaction of advanced glycation end products with their receptors/binding proteins. J Biol Chem, 269(13): 9889-9897.
  108. 108. Lander, H.M., Tauras, J.M., Ogiste, J.S., Hori, O., Moss, R.A. & Schmidt, A.M. 1997. Activation of the receptor for advanced glycation end products triggers a p21 ras-dependent mitogen-activated protein kinase pathway regulated by oxidant stress. J Biol Chem, 272(28): 17810-17814.
  109. 109. Goldin, A., Beckman, J.A., Schmidt, A.M. & Creager, M.A. 2006. Advanced glycation end products sparking the development of diabetic vascular injury. Circulation, 114(6): 597-605.
  110. 110. Cybulski MI, Gimbrone MA., 1991. Endothelial expression of a mononuclear leukocyte adhesion molecule during atherogenesis. Science. 251:788-791
  111. 111. Rubanyi, G.M. 1993. The role of endothelium in cardiovascular homeostasis and diseases. J Cardiovasc Pharmacol, 22: S15-S18.
  112. 112. Schiekofer, S., Andrassy, M., Chen, J., Rudofsky, G., Schneider, J., Wendt, T., Stefan, N., Humpert, P., Fritsche, A. & Stumvoll, M. 2003. Acute hyperglycemia causes intracellular formation of CML and activation of ras, p42/44 MAPK, and nuclear factor κB in PBMCs. Diabetes, 52(3): 621-633.
  113. 113. Barbosa, J.H., Oliveira, S.L. & Seara, L.T. 2008. The role of advanced glycation end-products (AGEs) in the development of vascular diabetic complications. Arq Bras Endocrinol Metabol, 52(6): 940-950.
  114. 114. Wautier, M., Chappey, O., Corda, S., Stern, D.M., Schmidt, A.M. & Wautier, J. 2001. Activation of NADPH oxidase by AGE links oxidant stress to altered gene expression via RAGE. Am J Physiol Endocrinol Metabol, 280(5): E685-E694.
  115. 115. Obrosova, I.G. 2002. How does glucose generate oxidative stress in peripheral nerve? Int Rev Neurobiol, 50: 3-35.
  116. 116. Jiang, J., Wang, Z. & Li, D. 2004. Effects of AGEs on oxidation stress and antioxidation abilities in cultured astrocytes. Biomed Environ Sc, 17(1): 79-86.
  117. 117. Yamagishi, S. & Matsui, T. 2010. Advanced glycation end products, oxidative stress and diabetic nephropathy. Oxid Med and Cell Longev, 3(2): 101-108.
  118. 118. Miura, J., Yamagishi, S., Uchigata, Y., Takeuchi, M., Yamamoto, H., Makita, Z. & Iwamoto, Y. 2003. Serum levels of non-carboxymethyllysine advanced glycation endproducts are correlated to severity of microvascular complications in patients with Type 1 diabetes. J Diabetes Complications, 17(1): 16-21.
  119. 119. Ono, Y., Aoki, S., Ohnishi, K., Yasuda, T., Kawano, K. & Tsukada, Y. 1998. Increased serum levels of advanced glycation end-products and diabetic complications. Diabetes Res Clin Prac, 41(2): 131-137.
  120. 120. Stitt, A.W. 2003. The role of advanced glycation in the pathogenesis of diabetic retinopathy. Exp Mol Pathol, 75(1): 95-108.
  121. 121. Chen, B., Jiang, D. & Tang, L. 2006. Advanced glycation end-products induce apoptosis involving the signaling pathways of oxidative stress in bovine retinal pericytes. Life Sci, 79(11): 1040-1048.
  122. 122. Kilhovd, B.K., Berg, T.J., Birkeland, K.I., Thorsby, P. & Hanssen, K.F. 1999. Serum levels of advanced glycation end products are increased in patients with type 2 diabetes and coronary heart disease. Diabetes Care, 22(9): 1543-1548.
  123. 123. Bowie, A., Owens, D., Collins, P., Johnson, A. & Tomkin, G.H. 1993. Glycosylated low density lipoprotein is more sensitive to oxidation: implications for the diabetic patient? Atherosclerosis, 102(1): 63-67.
  124. 124. Bucala, R., Makita, Z., Vega, G., Grundy, S., Koschinsky, T., Cerami, A. & Vlassara, H. 1994. Modification of low density lipoprotein by advanced glycation end products contributes to the dyslipidemia of diabetes and renal insufficiency. Proc Natl Acad Sci, 91(20): 9441-9445.
  125. 125. Sobenin, I., Tertov, V., Koschinsky, T., Bünting, C., Slavina, E., Dedovc, I. & Orekhov, A. 1993. Modified low density lipoprotein from diabetic patients causes cholesterol accumulation in human intimal aortic cells. Atherosclerosis, 100(1): 41-54.
  126. 126. Goh, S. & Cooper, M.E. 2008. The role of advanced glycation end products in progression and complications of diabetes. J Clin Endocrinol Metabol, 93(4): 1143-1152.
  127. 127. Eble, A.S., Thorpe, S.R. & Baynes, J.W. 1983. Nonenzymatic glucosylation and glucose-dependent cross-linking of protein. J Biol Chem, 258(15): 9406-9412.
  128. 128. Cooper, M.E., Bonnet, F., Oldfield, M. & Jandeleit-Dahm, K. 2001. Mechanisms of diabetic vasculopathy: an overview. Am J Hypertens, 14(5): 475-486.
  129. 129. Koya, D. & King, G.L. 1998. Protein kinase C activation and the development of diabetic complications. Diabetes, 47(6): 859-866.
  130. 130. Meier, M. & King, G.L. 2000. Protein kinase C activation and its pharmacological inhibition in vascular disease. Vasc Med, 5(3): 173-185.
  131. 131. Way, K., Katai, N. & King, G. 2001. Protein kinase C and the development of diabetic vascular complications. Diabet Med, 18(12): 945-959.
  132. 132. Inoguchi, T., Battan, R., Handler, E., Sportsman, J.R., Heath, W. & King, G.L. 1992. Preferential elevation of protein kinase C isoform beta II and diacylglycerol levels in the aorta and heart of diabetic rats: differential reversibility to glycemic control by islet cell transplantation. Proc Natl Acad Sci, 89(22): 11059-11063.
  133. 133. Shiba, T., Inoguchi, T., Sportsman, J.R., Heath, W.F., Bursell, S. & King, G.L. 1993. Correlation of diacylglycerol level and protein kinase C activity in rat retina to retinal circulation. Am J Physiol Endocrinol Metabol, 265(5): E783-E793.
  134. 134. Derubertis, F.R. & Craven, P.A. 1994. Activation of protein kinase C in glomerular cells in diabetes: mechanisms and potential links to the pathogenesis of diabetic glomerulopathy. Diabetes, 43(1): 1-8.
  135. 135. Thallas-Bonke, V., Thorpe, S.R., Coughlan, M.T., Fukami, K., Yap, F.Y., Sourris, K.C., Penfold, S.A., Bach, L.A., Cooper, M.E. & Forbes, J.M. 2008. Inhibition of NADPH oxidase prevents advanced glycation end product–mediated damage in diabetic nephropathy through a protein kinase C-α–dependent pathway. Diabetes, 57(2): 460-469.
  136. 136. Klann, E., Roberson, E.D., Knapp, L.T. et al., 1998. A role for superoxide in protein kinase C activation and induction of long-term potentiation. J Biol Chem. 273: 4516–4522.
  137. 137. Noh, H. & King, G. 2007. The role of protein kinase C activation in diabetic nephropathy. Kidney Int, 72(: S49-S53.
  138. 138. Jones, S., O'donnell, V., Wood, J., Broughton, J., Hughes, E. & Jones, O. 1996. Expression of phagocyte NADPH oxidase components in human endothelial cells. Am J Physiol Heart Circ Physiol, 271(4): H1626-H1634.
  139. 139. Pagano, P.J., Clark, J.K., Cifuentes-Pagano, M.E., Clark, S.M., Callis, G.M. & Quinn, M.T. 1997. Localization of a constitutively active, phagocyte-like NADPH oxidase in rabbit aortic adventitia: enhancement by angiotensin II. Proc Natl Acad Sci, 94(26): 14483-14488.
  140. 140. Greiber, S., Münzel, T., Kästner, S., Müller, B., Schollmeyer, P. & Pavenstädt, H. 1998. NAD (P) H oxidase activity in cultured human podocytes: effects of adenosine triphosphate. Kidney Int, 53(3): 654-663.
  141. 141. Patterson C, Ruef J, Madamanchi NR, Barry-Lane P, Hu Z, Horaist C, Ballinger CA, Brasier AR, Bode C, Runge MS, 1999: Stimulation of a vascular smooth muscle cell NAD(P)H oxidase by thrombin: evidence that p47phox may participate in forming this oxidase in vitro and in vivo. J Biol Chem. 274:19814–19822.
  142. 142. Hink, U., Li, H., Mollnau, H., Oelze, M., Matheis, E., Hartmann, M., Skatchkov, M., Thaiss, F., Stahl, R.A. & Warnholtz, A. 2001. Mechanisms underlying endothelial dysfunction in diabetes mellitus. Circ Res, 88(2): e14-e22.
  143. 143. Guzik, T.J., Mussa, S., Gastaldi, D., Sadowski, J., Ratnatunga, C., Pillai, R. & Channon, K.M. 2002. Mechanisms of increased vascular superoxide production in human diabetes mellitus Role of NAD (P) H oxidase and endothelial nitric oxide synthase. Circulation, 105(14): 1656-1662.
  144. 144. Kim, Y.K., Lee, M., Son, S.M., Kim, I.J., Lee, W.S., Rhim, B.Y., Hong, K.W. & Kim, C.D. 2002. Vascular NADH oxidase is involved in impaired endothelium-dependent vasodilation in OLETF rats, a model of type 2 diabetes. Diabetes, 51(2): 522-527.
  145. 145. Etoh, T., Kakimoto, M., Sonoda, N., Kobayashi, K., Kuroda, J., Sumimoto, H. & Nawata, H. 2003. Increased expression of NAD (P) H oxidase subunits, NOX4 and p22phox, in the kidney of streptozotocin-induced diabetic rats and its reversibity by interventive insulin treatment. Diabetologia, 46(10): 1428-1437.
  146. 146. Ellis, E., Grant, M.B., Murray, F.T., Wachowski, M.B., Guberski, D.L., Kubilis, P.S. & Lutty, G.A. 1998. Increased NADH oxidase activity in the retina of the BBZ/Wor diabetic rat. Free Radic Biol Med, 24(1): 111-120.
  147. 147. Ceriello, A., Ihnat, M.A. & Thorpe, J.E. 2009. The “metabolic memory”: is more than just tight glucose control necessary to prevent diabetic complications? J Clin Endocrinol Metabol, 94(2): 410-415.
  148. 148. Liu S, Lee I.M., Song Y, Van Denburgh M, Cook N.R., Manson J.E., Buring J.E., 2006. Vitamin E and risk of type 2 diabetes in the women’s health study randomized controlled trial. Diabetes, 55:2856–62.
  149. 149. Rhee, S., Jeong, Y. & Choi, J. 2005. Effects of vitamin E on phospholipase A2 activity and oxidative damage to the liver in streptozotocin-induced diabetic rats. Ann Nutr Metabol, 49(6): 392-396.
  150. 150. Devaraj, S., Leonard, S., Traber, M.G., Jialal, I., 2008. Gamma-tocopherol supplementation alone and in combination with alpha-tocopherol alters biomarkers of oxidative stress and inflammation in subjects with metabolic syndrome. Free Radic Biol Med. 44:1203–1208
  151. 151. Minamiyama, Y., Takemura, S., Bito, Y., Shinkawa, H., Tsukioka, T., Nakahira, A., Suehiro, S. & Okada, S. 2008. Supplementation of α-tocopherol improves cardiovascular risk factors via the insulin signalling pathway and reduction of mitochondrial reactive oxygen species in type II diabetic rats. Free Radic Res, 42(3): 261-271.
  152. 152. Davi, G., Ciabattoni, G., Consoli, A., Mezzetti, A., Falco, A., Santarone, S., Pennese, E., Vitacolonna, E., Bucciarelli, T., Costantini, F., Capani, F., Patrono, C., 1999. In vivo formation of 8-iso-prostaglandin f2alpha and platelet activation in diabetes mellitus: effects of improved metabolic control and vitamin E supplementation. Circulation. 99, 224–229
  153. 153. Wu, J.H., Ward, N.C., Indrawan, A.P., Almeida, C., Hodgson, J.M., Proudfoot, J.M., Puddey, I.B. & Croft, K.D. 2007. Effects of α-tocopherol and mixed tocopherol supplementation on markers of oxidative stress and inflammation in type 2 diabetes. Clin Chem, 53(3): 511-519.
  154. 154. Montero, A., Munger, K.A., Khan, R.Z., Valdivielso, J.M., Morrow, J.D., Guasch, A., Ziyadeh, F.N. & Badr, K.F. 2000. F2-isoprostanes mediate high glucose-induced TGF-&bgr; synthesis and glomerular proteinuria in experimental type I diabetes. Kidney Int, 58(5): 1963-1972.
  155. 155. Je, H., Shin, C., Park, H., Huh, I. & Sohn, U. 2001. The comparison of vitamin C and vitamin E on the protein oxidation of diabetic rats. J Auton Pharmacol, 21(5): 231-236.
  156. 156. Schleicher, E.D., Wagner, E. & Nerlich, A.G. 1997. Increased accumulation of the glycoxidation product N (epsilon)-(carboxymethyl) lysine in human tissues in diabetes and aging. J Clin Investig, 99(3): 457.
  157. 157. Jialal, I., Devaraj, S. & Venugopal, S. 2002. Oxidative stress, inflammation, and diabetic vasculopathies: the role of alpha tocopherol therapy. Free Rad Res, 36(12): 1331-1336.
  158. 158. Sakamoto, W., Fujie, K., Handa, H., Ogihara, T. & Mino, M. 1989. In vivo inhibition of superoxide production and protein kinase C activity in macrophages from vitamin E-treated rats. Int J Vitam Nutr Res, 60(4): 338-342.
  159. 159. Kunisaki, M., Bursell, S., Clermont, A.C., Ishii, H., Ballas, L.M., Jirousek, M.R., Umeda, F., Nawata, H. & King, G.L. 1995. Vitamin E prevents diabetes-induced abnormal retinal blood flow via the diacylglycerol-protein kinase C pathway. Am J Physiol Endocrinol Metabol, 269(2): E239-E246.
  160. 160. Koya, D., Lee, I., Ishii, H., Kanoh, H. & King, G.L. 1997. Prevention of glomerular dysfunction in diabetic rats by treatment with d-alpha-tocopherol. Clin J Am Soc Nephrol, 8(3): 426-435.
  161. 161. Yülek, F., Or, M., Özoğul, C., Isik, A.C., Ari, N., Stefek, M., Bauer, V. & Karasu, C. 2007. Effects of stobadine and vitamin E in diabetes-induced retinal abnormalities: involvement of oxidative stress. Arch Med Sc, 38(5): 503-511.
  162. 162. Hamblin M, Smith HM, Hill MF., 2007. Dietary supplementation with vitamin E ameliorates cardiac failure in type I diabetic cardiomyopathy by suppressing myocardial generation of 8-iso-prostaglandin F2-alpha and oxidized glutathione. J Card Fail, 13(10): 884-892.
  163. 163. Boaz, M., Smetana, S., Weinstein, T., Matas, Z., Gafter, U., Iaina, A., Knecht, A., Weissgarten, Y., Brunner, D. & Fainaru, M. 2000. Secondary prevention with antioxidants of cardiovascular disease in endstage renal disease (SPACE): randomised placebo-controlled trial. The Lancet, 356(9237): 1213-1218.
  164. 164. Skyrme-Jones, R.A.P., O’Brien, R.C., Berry, K.L. & Meredith, I.T. 2000. Vitamin E supplementation improves endothelial function in type I diabetes mellitus: a randomized, placebo-controlled study. J Am Coll Cardiol, 36(1): 94-102.
  165. 165. Heart Outcomes Prevention Evaluation (HOPE) Study Investigators, 2000. Effects of ramipril on cardiovascular and microvascular outcomes in people with diabetes mellitus: results of the HOPE study and MICRO-HOPE substudy. Lancet. 355: 253-259.
  166. 166. Lonn E, Yusuf S, Hoogwerf B, Pogue J, Yi Q, Zinman B, Bosch J, Dagenais G, Mann JFE, Gerstein HC, 2002. Effects of vitamin E on cardiovascular and microvascular outcomes in high-risk patients with diabetes: Results of the HOPE study and MICRO-HOPE substudy. Diabetes Care. 25:1919-1927.
  167. 167. Lee, E.Y., Lee, M.Y., Hong, S.W., Chung, C.H. & Hong, S.Y. 2007. Blockade of oxidative stress by vitamin C ameliorates albuminuria and renal sclerosis in experimental diabetic rats. Yonsei Med J, 48(5): 847-855.
  168. 168. Sridulyakul, P., Wongeak-in, N. & Patumraj, S. 2010. Increased nitric oxide level in diabetic rats from vitamin C supplementation: an in vivo detection using diaminofluorescein. Asian Biomed, 2(5): 371-379.
  169. 169. Eriksson, J. & Kohvakka, A. 1995. Magnesium and ascorbic acid supplementation in diabetes mellitus. Ann Nutr Metabol, 39(4): 217-223.
  170. 170. Paolisso, G., Balbi, V., Volpe, C., Varricchio, G., Gambardella, A., Saccomanno, F., Ammendola, S., Varricchio, M. & D'Onofrio, F. 1995. Metabolic benefits deriving from chronic vitamin C supplementation in aged non-insulin dependent diabetics. J Am Coll Nutr, 14(4): 387-392.
  171. 171. Mullan, B.A., Young, I.S., Fee, H. & McCance, D.R. 2002. Ascorbic acid reduces blood pressure and arterial stiffness in type 2 diabetes. Hypertension, 40(6): 804-809.
  172. 172. Regensteiner, J.G., Popylisen, S., Bauer, T.A., Lindenfeld, J., Gill, E., Smith, S., Oliver-Pickett, C.K., Reusch, J.E. & Weil, J.V. 2003. Oral L-arginine and vitamins E and C improve endothelial function in women with type 2 diabetes. Vasc Med, 8(3): 169-175.
  173. 173. Mazloom, Z., Hejazi, N., Dabbaghmanesh, M.H., Tabatabaei, H.R., Ahmadi, A. & Ansar, H. 2011. Effect of vitamin C supplementation on postprandial oxidative stress and lipid profile in type 2 diabetic patients. Pakistan J Biol Sci: PJBS, 14(19): 900-904.
  174. 174. Ting, H.H., Timimi, F.K., Boles, K.S., Creager, S.J., Ganz, P. & Creager, M.A. 1996. Vitamin C improves endothelium-dependent vasodilation in patients with non-insulin-dependent diabetes mellitus. J Clin Investig, 97(1): 22.
  175. 175. Timimi, F.K., Ting, H.H., Haley, E.A., Roddy, M., Ganz, P. & Creager, M.A. 1998. Vitamin C improves endothelium-dependent vasodilation in patients with insulin-dependent diabetes mellitus. J Am Coll Cardiol, 31(3): 552-557.
  176. 176. Chen, H., Karne, R.J., Hall, G., Campia, U., Panza, J.A., Cannon, R.O., Wang, Y., Katz, A., Levine, M. & Quon, M.J. 2006. High-dose oral vitamin C partially replenishes vitamin C levels in patients with type 2 diabetes and low vitamin C levels but does not improve endothelial dysfunction or insulin resistance. Am J Physiol Heart Circ Physiol, 290(1): H137-H145.
  177. 177. Jackson, T.S., Xu, A., Vita, J.A. & Keaney, J.F. 1998. Ascorbate prevents the interaction of superoxide and nitric oxide only at very high physiological concentrations. Circ Res, 83(9): 916-922.
  178. 178. Sherman, D.L., Keaney, J.F., Biegelsen, E.S., Duffy, S.J., Coffman, J.D. & Vita, J.A. 2000. Pharmacological concentrations of ascorbic acid are required for the beneficial effect on endothelial vasomotor function in hypertension. Hypertension, 35(4): 936-941.
  179. 179. Darko, D., Dornhorst, A., Kelly, F., Ritter, J. & Chowienczyk, P. 2002. Lack of effect of oral vitamin C on blood pressure, oxidative stress and endothelial function in Type II diabetes. Clin Sci, 103(4): 339-344.
  180. 180. Corradini, E., Foglia, P., Giansanti, P., Gubbiotti, R., Samperi, R. & Lagana, A. 2011. Flavonoids: chemical properties and analytical methodologies of identification and quantitation in foods and plants. Nat Prod Res, 25(5): 469-495.
  181. 181. Middleton, E., Kandaswami, C. & Theoharides, T.C. 2000. The effects of plant flavonoids on mammalian cells: implications for inflammation, heart disease, and cancer. Pharmacol Rev, 52(4): 673-751.
  182. 182. Batra P and Sharma AK, 2013 Anti-cancer potential of flavonoids: recent trends and future perspectives 3 Biotechnology DOI 10.1007/s13205-013-0117-5
  183. 183. Selway, J.W. 1986. Antiviral activity of flavones and flavans. Progr Clin Biol Res, 213: 521-536.
  184. 184. Rice-Evans, C.A., Miller, N.J. & Paganga, G. 1996. Structure-antioxidant activity relationships of flavonoids and phenolic acids. Free Radic Biol Med, 20(7): 933-956.
  185. 185. Heim, K. E.; Tagliaferro, A. R.; Bobilya D. J. Flavonoid antioxidants: chemistry, metabolism and sructure-activity relationships. J Nutr Biochem. 2002, 13, 572-584
  186. 186. Groot, H.d. & Rauen, U. 1998. Tissue injury by reactive oxygen species and the protective effects of flavonoids. Fund Clin Pharmacol, 12(3): 249-255.
  187. 187. Narayana K.R., Reddy M.S., Chaluvadi M.R., Krishna D.R., 2001. Bioflavonoids classification, pharmacological, biochemical effects and therapeutic potential. Indian J Physiol Pharmacol; 33: 2-16.
  188. 188. Wu, C. & Yen, G. 2005. Inhibitory effect of naturally occurring flavonoids on the formation of advanced glycation endproducts. J Agr Food Chem, 53(8): 3167-3173.
  189. 189. Yamabe N, Yokozawa T, Oya T and Kim M, 2006. Therapeutic potential of (-) epigallocatechin 3-Ogallate on renal damage in diabetic nephropathy model rats. J Pharmacol Exp Therapeut, 319: 228-236.
  190. 190. Urios, P., Grigorova-Borsos, A. & Sternberg, M. 2007. Flavonoids inhibit the formation of the cross-linking AGE pentosidine in collagen incubated with glucose, according to their structure. Eur J Nutr, 46(3): 139-146.
  191. 191. Kim, H.Y., Lee, J.M., Yokozawa, T., Sakata, K. & Lee, S. 2011. Protective activity of flavonoid and flavonoid glycosides against glucose-mediated protein damage. Food Chem, 126(3): 892-895.
  192. 192. Goodarzi, M., Zal, F., Malakooti, M. & Sadeghian, M.S.S. 2006. Inhibitory activity of flavonoids on the lens aldose reductase of healthy and diabetic rats. Acta Medica Iranica, 44(1).
  193. 193. Jung, S.H., Lee, Y.S., Lee, S., Lim, S.S., Kim, Y.S. & Shin, K.H. 2002. Isoflavonoids from the rhizomes of Belamcanda chinensis and their effects on aldose reductase and sorbitol accumulation in streptozotocin induced diabetic rat tissues. Arch Pharm Res, 25(3): 306-312.
  194. 194. Kim, J., Kim, J.H., Jun, H., Yu, Y.S. & Kim, K. 2010. Inhibition of protein kinase C δ attenuates blood-retinal barrier breakdown in diabetic retinopathy. Am J Clin Pathol, 176(3): 1517-1524.
  195. 195. Frank, R.N., 2004. Diabetic retinopathy. New Engl J Med. 350: 48–58
  196. 196. Kumar, B., Gupta, S.K., Srinivasan, B., Nag, T.C., Srivastava, S. & Saxena, R. 2012. Hesperetin ameliorates hyperglycemia induced retinal vasculopathy via anti-angiogenic effects in experimental diabetic rats. Vasc Pharmacol, 57(5): 201-207.
  197. 197. Kandhare, A.D., Raygude, K.S., Ghosh, P., Ghule, A.E. & Bodhankar, S.L. 2012. Neuroprotective effect of naringin by modulation of endogenous biomarkers in streptozotocin induced painful diabetic neuropathy. Fitoterapia, 83(4): 650-659.
  198. 198. Valsecchi, A.E., Franchi, S., Panerai, A.E., Rossi, A., Sacerdote, P. & Colleoni, M. 2011. The soy isoflavone genistein reverses oxidative and inflammatory state, neuropathic pain, neurotrophic and vasculature deficits in diabetes mouse model. Eur J Clin Pharmacol, 650(2): 694-702.
  199. 199. Wang, G., Li, W., Lu, X., Bao, P. & Zhao, X. 2012. Luteolin ameliorates cardiac failure in type I diabetic cardiomyopathy. J Diabetes Complications, 26(4): 259-265.
  200. 200. Manuel y Keenoy, B., Vertommen, J. & De Leeuw, I. 1999. The effect of flavonoid treatment on the glycation and antioxidant status in Type 1 diabetic patients. Diabetes, Nutr Metabol, 12(4): 256-263.
  201. 201. Pandey, K.B., Mishra, N. & Rizvi, S.I. 2009. Myricetin may provide protection against oxidative stress in type 2 diabetic erythrocytes. Int J Biosci, 64(9): 626.
  202. 202. Kamalakkannan, N. & Prince, P.S.M. 2006. Antihyperglycaemic and Antioxidant Effect of Rutin, a Polyphenolic Flavonoid, in Streptozotocin‐Induced Diabetic Wistar Rats. Basic Clin Pharmacol Toxicol, 98(1): 97-103.
  203. 203. Sattanathan, K., Dhanapal, C., Umarani, R. & Manavalan, R. 2011. Beneficial health effects of rutin supplementation in patients with diabetes mellitus. JAPS, 1(08): 227-231.
  204. 204. Hao, H., Shao, Z., Tang, D., Lu, Q., Chen, X., Yin, X., Wu, J. & Chen, H. 2012. Preventive effects of rutin on the development of experimental diabetic nephropathy in rats. Life Sci.
  205. 205. Cervantes-Laurean, D.D. Schramm, E.L. Jacobson, I. Halaweish, G.G. Bruckner, G.A., 2006. Boissonneault. Inhibition of advanced glycation end product formation on collagen by rutin and its metabolites. J Nutr Biochem. 17: 531–540
  206. 206. Campanero, M.A., Escolar, M., Perez, G., Garcia-Quetglas, E., Sadaba, B. & Azanza, J.R. 2010. Simultaneous determination of diosmin and diosmetin in human plasma by ion trap liquid chromatography–atmospheric pressure chemical ionization tandem mass spectrometry: Application to a clinical pharmacokinetic study. J Pharmaceut Biomed Anal, 51(4): 875-881.
  207. 207. Srinivasan S, Pari L, 2012. Ameliorative effect of diosmin, a citrus flavonoid against streptozotocin-nicotinamide generated oxidative stress induced diabetic rats. Chem Biol Interact. 195: 43–51
  208. 208. Tang, D., Wei, Y., Gao, Y., Yin, X., Yang, D., Mou, J. & Jiang, X. 2011. Protective effects of rutin on rat glomerular mesangial cells cultured in high glucose conditions. Phytother Res, 25(11): 1640-1647.
  209. 209. Machha, A., Achike, F.I., Mustafa, A.M. & Mustafa, M.R. 2007. Quercetin, a flavonoid antioxidant, modulates endothelium-derived nitric oxide bioavailability in diabetic rat aortas. Nitric Oxide, 16(4): 442-447.
  210. 210. Alkhamees, O.A. 2013. Morin a Flavonoid Exerts Antioxidant Potential in Streptozotocin-induced Hepatotoxicity. Br J Pharmacol Toxicol 4(1): 10-17, 2013
  211. 211. Kapoor, R. & Kakkar, P. 2012. Protective role of morin, a flavonoid, against high glucose induced oxidative stress mediated apoptosis in primary Rat hepatocytes. PloS one, 7(8): e41663.
  212. 212. Coskun, O., Kanter, M., Korkmaz, A. & Oter, S. 2005. Quercetin, a flavonoid antioxidant, prevents and protects streptozotocin-induced oxidative stress and β-cell damage in rat pancreas. Pharmacol Res, 51(2): 117-123.
  213. 213. Anjaneyulu, M. & Chopra, K. 2004. Quercetin, an anti‐oxidant bioflavonoid, attenuates diabetic nephropathy in rats. Clin Exp Pharmacol Physiol, 31(4): 244-248.
  214. 214. Mahmoud AM, Ashour MB, Abdel-Moneim A, Ahmed OM, 2012. Hesperidin and naringin attenuate hyperglycemia-mediated oxidative stress and proinflammatory cytokine production in high fat fed/streptozotocin-induced type 2 diabetic rats. J Diabetes Complications. 26: 483-490.
  215. 215. Shi, X., Liao, S., Mi, H., Guo, C., Qi, D., Li, F., Zhang, C. & Yang, Z. 2012. Hesperidin Prevents Retinal and Plasma Abnormalities in Streptozotocin-Induced Diabetic Rats. Molecules, 17(11): 12868-12881.
  216. 216. Annadurai, T., Muralidharan, A.R., Joseph, T., Hsu, M., Thomas, P. & Geraldine, P. 2012. Antihyperglycemic and antioxidant effects of a flavanone, naringenin, in streptozotocin–nicotinamide-induced experimental diabetic rats. J Physiol Biochem, 68(3): 307-318.
  217. 217. Tsai, S., Huang, C., Mong, M., Kam, W., Huang, H. & Yin, M. 2011. Anti-inflammatory and antifibrotic effects of naringenin in diabetic mice. J Agr Food Chem, 60(1): 514-521.
  218. 218. Soto, C., Pérez, J., García, V., Uría, E., Vadillo, M. & Raya, L. 2010. Effect of silymarin on kidneys of rats suffering from alloxan-induced diabetes mellitus. Phytomedicine, 17(14): 1090-1094.
  219. 219. Wang, G.G., Lu, X.H., Li, W., Zhao, X. & Zhang, C. 2011. Protective effects of luteolin on diabetic nephropathy in STZ-induced diabetic rats. Evid Base Compl Alternative Med, article ID 323171, 7 pages.
  220. 220. Qian, L., Wang, H., Chen, Y., Chen, F., Ma, Y., Bruce, I.C. & Xia, Q. 2010. Luteolin reduces high glucose-mediated impairment of endothelium-dependent relaxation in rat aorta by reducing oxidative stress. Pharmacol Res, 61(4): 281-287.
  221. 221. El‐Bassossy, H.M., Abo‐Warda, S.M. & Fahmy, A. 2013. Chrysin and Luteolin Attenuate Diabetes‐Induced Impairment in Endothelial‐Dependent Relaxation: Effect on Lipid Profile, AGEs and NO Generation. Phytother Res. doi: 10.1002/ptr.4917.
  222. 222. Panda, S. & Kar, A. 2007. Apigenin (4 ‘, 5, 7‐trihydroxyflavone) regulates hyperglycaemia, thyroid dysfunction and lipid peroxidation in alloxan‐induced diabetic mice. J Pharm Pharmacol, 59(11): 1543-1548.
  223. 223. Ihm, S., Lee, J., Kim, S., Seung, K., Schini-Kerth, V.B., Chang, K. & Oak, M. 2009. Catechin prevents endothelial dysfunction in the prediabetic stage of OLETF rats by reducing vascular NADPH oxidase activity and expression. Atherosclerosis, 206(1): 47-53.
  224. 224. Fu, Z., Yuskavage, J. & Liu, D. 2013. Dietary Flavonol Epicatechin Prevents the Onset of Type 1 Diabetes in Nonobese Diabetic Mice. J Agr Food Chem, 61(18): 4303-4309.
  225. 225. Kim, M.J. & Lim, Y. 2013. Protective Effect of Short-Term Genistein Supplementation on the Early Stage in Diabetes-Induced Renal Damage. Mediat Inflamm, 2013: 1-14
  226. 226. Roghani, M., Vaez Mahdavi, M., Jalali‐Nadoushan, M., Baluchnejadmojarad, T., Naderi, G., Roghani‐Dehkordi, F., Taghi Joghataei, M. & Kord, M. 2013. Chronic Administration of Daidzein, a Soybean Isoflavone, Improves Endothelial Dysfunction and Attenuates Oxidative Stress in Streptozotocin‐induced Diabetic Rats. Phytother Res, 27(1): 112-117.
  227. 227. Fukui, T., Noma, T., Mizushige, K., Aki, Y., Kimura, S. & Abe, Y. 2000. Dietary troglitazone decreases oxidative stress in early stage type II diabetic rats. Life Sci, 66(21): 2043-2049.
  228. 228. Dobsak P, Courderot-Masuyer C, Siegelova J, Svacinova H, Jancik J, Vergely-Vanriessen C, Rochette, 2001. Antioxidant properties of aminoguanidine: a paramagnetic resonance test. Scripta Med (BRNO) 74: 45–50.
  229. 229. Elmalí, E., Altan, N. & Bukan, N. 2004. Effect of the sulphonylurea glibenclamide on liver and kidney antioxidant enzymes in streptozocin-induced diabetic rats. Drugs in R & D, 5(4): 203-208
  230. 230. Haendeler, J., Hoffmann, J., Zeiher, A.M. & Dimmeler, S. 2004. Antioxidant effects of statins via s-nitrosylation and activation of thioredoxin in endothelial cells a novel vasculoprotective function of statins. Circulation, 110(7): 856-861.
  231. 231. Gumieniczek, A., Hopkała, H., Roliński, J. & Bojarska-Junak, A. 2005. Antioxidative and anti-inflammatory effects of repaglinide in plasma of diabetic animals. Pharmacol Res, 52(2): 162-166.

Written By

Omolola R. Ayepola, Nicole L. Brooks and Oluwafemi O. Oguntibeju

Submitted: 16 September 2013 Published: 05 February 2014