Next Article in Journal
Effects of Antioxidant in Adjunct with Periodontal Therapy in Patients with Type 2 Diabetes: A Systematic Review and Meta-Analysis
Previous Article in Journal
Early Cytokine-Induced Transient NOX2 Activity Is ER Stress-Dependent and Impacts β-Cell Function and Survival
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Pathophysiological Association between Diabetes Mellitus and Endothelial Dysfunction

by
Tatsuya Maruhashi
1 and
Yukihito Higashi
1,2,*
1
Department of Cardiovascular Regeneration and Medicine, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima 734-8553, Japan
2
Division of Regeneration and Medicine, Medical Center for Translational and Clinical Research, Hiroshima University Hospital, Hiroshima 734-8551, Japan
*
Author to whom correspondence should be addressed.
Antioxidants 2021, 10(8), 1306; https://doi.org/10.3390/antiox10081306
Submission received: 5 July 2021 / Revised: 17 August 2021 / Accepted: 17 August 2021 / Published: 18 August 2021

Abstract

:
Endothelial dysfunction plays a critical role in atherosclerosis progression, leading to cardiovascular complications. There are significant associations between diabetes mellitus, oxidative stress, and endothelial dysfunction. Oxidative stress is increased by chronic hyperglycemia and acute glucose fluctuations induced by postprandial hyperglycemia in patients with diabetes mellitus. In addition, selective insulin resistance in the phosphoinositide 3-kinase/Akt/endothelial nitric oxide (NO) synthase pathway in endothelial cells is involved in decreased NO production and increased endothelin-1 production from the endothelium, resulting in endothelial dysfunction. In a clinical setting, selecting an appropriate therapeutic intervention that improves or augments endothelial function is important for preventing diabetic vascular complications. Hypoglycemic drugs that reduce glucose fluctuations by decreasing the postprandial rise in blood glucose levels, such as glinides, α-glucosidase inhibitors and dipeptidyl peptidase 4 inhibitors, and hypoglycemic drugs that ameliorate insulin sensitivity, such as thiazolidinediones and metformin, are expected to improve or augment endothelial function in patients with diabetes. Glucagon-like peptide 1 receptor agonists, metformin, and sodium-glucose cotransporter 2 inhibitors may improve endothelial function through multiple mechanisms, some of which are independent of glucose control or insulin signaling. Oral administration of antioxidants is not recommended in patients with diabetes due to the lack of evidence for the efficacy against diabetic complications.

1. Introduction

The number of patients with diabetes has been increasing worldwide. A pooled analysis showed that the prevalence of diabetes in adults has been increasing, and it was estimated that the number of adults with diabetes in the world increased from 108 million in 1980 to 422 million in 2014 [1]. Diabetic patients are at high risk for developing microvascular and macrovascular complications. Diabetic vascular complications are major causes of morbidity and mortality in diabetic patients. Microvascular complications such as neuropathy, nephropathy, and retinopathy are major causes of decreased quality of life. Macrovascular complications including coronary heart disease, cerebrovascular disease, and peripheral artery disease, are major causes of mortality. A systematic literature review has shown that cardiovascular disease affects approximately 32.2% of patients with type 2 diabetes (21.1% with coronary heart disease, 14.6% with angina, 10.0% with myocardial infarction, and 7.6% with stroke) and that cardiovascular disease is the cause of death in 9.9% of patients with type 2 diabetes aged 63.6 ± 6.9 years, accounting for 50.3% of all deaths in diabetic patients [2]. Therefore, inhibiting the progression of atherosclerosis and preventing the development of diabetic vascular complications is clinically important for a better prognosis in diabetic patients.
Endothelial dysfunction is not only an early event of atherosclerosis but also plays a critical role in atherosclerosis progression, leading to the development of vascular complications [3,4,5]. Diabetes mellitus is associated with endothelial dysfunction [6,7,8,9]. Therefore, it is important to understand the mechanisms underlying endothelial dysfunction caused by diabetes mellitus and to select treatments that improve or augment endothelial function for preventing diabetic vascular complications. In this review, we discuss the current understanding of the mechanisms of endothelial dysfunction and recommended therapeutic options for improving endothelial function in patients with diabetes mellitus.

2. The Resting Endothelium

The vascular endothelium acts as an endocrine organ that secrets a variety of vasoactive agents: vasodilators such as prostaglandin I2, endothelium-derived hyperpolarizing factor, and nitric oxide (NO), and vasoconstrictors such as angiotensin II, thromboxane A2, and endothelin-1 (ET-1) [10]. The resting endothelium acts as a gatekeeper that maintains vascular homeostasis by regulating the moment-to-moment balance between vasodilation and vasoconstriction, antithrombosis and prothrombosis, anti-inflammation and proinflammation, antioxidation and pro-oxidation, and vascular smooth muscle cell growth inhibition and growth promotion [3]. Endothelial dysfunction refers to the inability of endothelial cells to maintain vascular homeostasis due to the disturbed balance between endothelium-derived proatherosclerotic factors and antiatherosclerotic factors in favor of proatherosclerotic factors, leading to the initiation and progression of atherosclerosis. NO released from the endothelium has a variety of antiatherosclerotic effects such as vasodilation, inhibition of the proliferation of vascular smooth muscle cells, inhibition of leukocyte adhesion, and inhibition of platelet adhesion and aggregation. Therefore, endothelial dysfunction often refers to a condition in which increased NO inactivation and/or decreased NO production from the endothelium results in reduced NO bioavailability.
Diabetes mellitus is associated with endothelial dysfunction [11,12,13]. Clinical studies have shown that endothelial function assessed by endothelium-dependent vasodilation is impaired in diabetic patients [14,15]. Although the pathogenesis has not been fully elucidated, several mechanisms underlying the relationship between diabetes mellitus and endothelial dysfunction have been proposed.

3. Mechanisms Underlying Endothelial Dysfunction in Diabetes Mellitus

3.1. Oxidative Stress

Reactive oxygen species (ROS) are derived from molecular oxygen. Oxidative stress refers to the disturbed balance between the antioxidant system and ROS in favor of ROS. When the counteracting effect of the antioxidant system is insufficient, harmful effects of ROS such as inhibition of signal transduction pathways or normal cellular functions through damage to cellular lipids, proteins, or DNA become evident. In human cells, ROS are produced by various enzymatic sources such as nicotinamide-adenine dinucleotide phosphate (NADPH) oxidases, xanthine dehydrogenase/oxidase, the mitochondrial electron transport chain, uncoupled endothelial NO synthase (eNOS), cyclooxygenase, lipoxygenase, and glucose oxidase [16]. ROS include free radical species such as superoxide anion radical (O2•−), peroxyl radical, alkoxyl radical, and hydroxyl radical, and non-radical species such as singlet molecular oxygen, hydrogen peroxide, organic hydroperoxides, hypochlorous acid, and ozone [16].
There is an interaction between endothelial function and oxidative stress [3,17]. O2•−, one of the free radical species, is produced through the removal of one electron from molecular oxygen. NO is directly inactivated by O2•− with high affinity, resulting in decreased NO bioavailability. In addition, peroxynitrite is produced as a result of the direct reaction between NO and O2•− [18]. Peroxynitrite is a highly potent oxidant that can cause lipid peroxidation, protein tyrosine nitration, DNA damage, and cell death [18]. Tetrahydrobiopterin (BH4), an essential eNOS cofactor, is oxidized by peroxynitrite to the biologically inactive form, resulting in reduced BH4 availability. Under a condition of insufficient BH4, O2•− is produced instead of NO from uncoupled eNOS [19]. Therefore, O2•− is closely associated with the development of endothelial dysfunction. Once an oxidative condition is established, endothelial function continues to be impaired through a vicious cycle of increased O2•− and decreased NO bioavailability. In diabetes mellitus, chronic hyperglycemia, acute glucose fluctuations, and insulin resistance are regarded as major causes of endothelial dysfunction.

3.2. Chronic Hyperglycemia and Oxidative Stress

Intracellular O2•− is mainly produced from mitochondria [20]. Pyruvate is generated by glycolysis in the cytosol and is used for ATP synthesis by oxidative phosphorylation in mitochondria. After the transportation of pyruvate into the mitochondria, pyruvate is oxidized by the tricarboxylic acid (TCA) cycle to produce H2O, CO2, nicotinamide adenine dinucleotide (NADH), and 1,5-dihydro-flavin adenine dinucleotide (FADH2) [21]. Electrons from mitochondrial NADH and FADH2 are used as energy for ATP synthesis by the electron-transport chain at the inner mitochondrial membrane. Electrons from NADH and FADH2 are transferred through the mitochondrial electron-transport chain, which is coupled with proton pumping by the electron-transport chain from the mitochondrial matrix into the intermembrane space. As a result of proton pumping, a proton gradient is generated across the inner membrane of mitochondria, which provides the energy for driving the ATP synthase. In a hyperglycemic state, increased production of NADH and FADH2 by the TCA cycle leads to increased transportation of NADH and FADH2 to the electron-transport chain. Since NADH and FADH2 serve as electron donors, electron transfer and proton pumping through the electron-transport chain is concomitantly enhanced and a proton gradient across the inner mitochondrial membrane is increased. As a result, electron transfer and proton pumping are inhibited, resulting in an increase in electron leak from the electron-transport chain and a subsequent increase in O2•− generation in mitochondria (Figure 1) [21].
GAPDH is the glycolytic enzyme that is essential for maintaining glycolysis. Hyperglycemia-induced overproduction of mitochondrial O2•− partially inhibits the activity of GAPDH. Therefore, the inhibition of GAPDH activity by mitochondrial O2•− causes the accumulation of glycolytic metabolites upstream of GAPDH and the increased flux of upstream metabolites into pathways of glucose overutilization (Figure 1) [21].
An increase in glucose flux into the polyol pathway leads to increased consumption of NADPH. NADPH is required for the regeneration of reduced glutathione. Therefore, intracellular concentrations of reduced glutathione are decreased as a result of increased consumption of NADPH by an increase in glucose flux into the polyol pathway. Since reduced glutathione is a main intracellular antioxidant, intracellular oxidative stress is enhanced, leading to endothelial dysfunction.
Increased glucose flux into the hexosamine pathway may result in endothelial dysfunction. Fructose-6-phosphate is converted to glucosamine-6-phosphate in the hexosamine pathway, resulting in an increase in UDP-N-acetylglucosamine, which is required for reactions such as proteoglycan synthesis and O-linked glycoproteins formation. As a result of increased UDP-N-acetylglucosamine, transcriptional factors, nuclear proteins, and cytoplasmic proteins are modified by O-linked N-acetylglucosamine, leading to many alterations in both gene and protein functions. For instance, eNOS activity is inhibited by O-acetylglucosaminylation of the eNOS protein at the Akt site [22], leading to decreased NO production and consequent endothelial dysfunction.
Hyperglycemia-induced activation of protein kinase C (PKC) through an increase in diacylglycerol has a number of pathogenic effects such as decreased eNOS expression, increased ET-1 expression, increased plasminogen activator inhibitor-1 expression, increased transforming growth factor-β expression, NF-kB activation, and NADPH oxidase activation, leading to endothelial dysfunction.
Increased intracellular production of advanced glycation end products (AGE) precursors causes modifications of plasma proteins and extracellular matrix proteins and functional alterations of intracellular proteins. ROS production and NF-kB activation are induced by the activation of the receptor for AGEs on the surface of endothelial cells, leading to endothelial dysfunction. Therefore, endothelial dysfunction is caused by hyperglycemia-induced overproduction of mitochondrial O2•− and diversion of glycolytic flux from the normal glycolytic pathway to alternative metabolic pathways due to inhibition of GAPDH activity by mitochondrial O2•−.

3.3. Acute Glucose Fluctuations and Oxidative Stress

In healthy subjects, plasma glucose levels are controlled and maintained within a narrow range, whereas blood glucose levels are rapidly and greatly increased in the postprandial phase in patients with diabetes mellitus. Experimental and clinical studies have indicated that endothelial function is impaired by postprandial acute hyperglycemia through increased oxidative stress in patients with diabetes mellitus (Figure 1) [23]. Compared with constant high glucose, intermittent high glucose may have more harmful effects on endothelial cells. In vitro studies have shown that more apoptosis of endothelial cells was induced by intermittent high glucose through PKC activation and NADPH oxidase activation than by constant high glucose [24,25]. In addition, clinical studies have indicated that endothelial function is impaired by glucose fluctuations through oxidative stress. Monnier et al. reported that a marker of glucose fluctuations was strongly correlated with a marker of oxidative stress, whereas there was no significant correlation between the oxidative stress marker and any other glycemic parameters such as fasting plasma glucose and glycated hemoglobin (HbA1c) [26]. Torimoto et al. reported that the marker of glucose fluctuations was negatively correlated with a marker of endothelial function [27]. These findings suggest that endothelial function is impaired by glucose fluctuations through increased oxidative stress. Although the precise molecular mechanisms underlying the association between glucose fluctuations and enhanced oxidative stress have not been fully elucidated, it is postulated that cellular metabolic adaptation to toxic effects induced by high glucose may be facilitated by a constant high glucose concentration through constant feedback. In contrast, intermittent high glucose may fail to facilitate such adaptations to toxic effects induced by high glucose because of the lack of constant feedback, leading to higher oxidative stress and consequent endothelial dysfunction. HbA1c has been used as a therapeutic marker of glucose control. HbA1c, however, reflects time-averaged glucose exposure but not glucose fluctuations. Large clinical randomized studies in which HbA1c was used as a marker of glycemic control failed to demonstrate the superiority of intensive glucose control for prevention of cardiovascular events [28,29,30]. Therefore, it is necessary to pay attention not only to HbA1c and fasting plasma glucose levels but also to postprandial glucose levels for protecting the endothelium from oxidative injury caused by postprandial hyperglycemia.

3.4. Selective Insulin Resistance-Induced Endothelial Dysfunction

Insulin stimulates NO production in endothelial cells. Insulin receptor substrate (IRS) is phosphorylated by the binding of insulin to its cognate receptor on endothelial cells, resulting in the activation of the phosphoinositide 3-kinase (PI3-kinase)/Akt pathway. eNOS is phosphorylated at Ser1177 by Akt, resulting in increased NO production [31,32]. Insulin also stimulates ET-1 production in endothelial cells through activating the mitogen-activated protein kinase (MAPK)/extracellular signal-regulated kinase (ERK) pathway independent of the PI3-kinase/Akt/eNOS pathway [33]. The PI3-kinase/Akt/eNOS pathway is selectively impaired under the condition of insulin resistance because of reduced IRS expression in endothelial cells. In contrast, the MAPK/ERK/ET-1 pathway remains unchanged and is preferentially stimulated because of the compensatory hyperinsulinemia, leading to endothelial dysfunction (Figure 1) [34]. This phenomenon is referred to as selective insulin resistance.

3.5. Endothelial Dysfunction in Type 1 Diabetes

Endothelial function has been shown to be impaired in patients with type 1 diabetes [15,35]. Oxidative stress is increased and antioxidant defense is impaired in patients with type 1 diabetes [36,37,38], suggesting that oxidative stress is involved in endothelial dysfunction in patients with type 1 diabetes. Clinical studies have indicated that chronic hyperglycemia and acute hyperglycemia are associated with endothelial dysfunction in patients with type 1 diabetes [39,40,41]. In addition, experimental studies have indicated the possibility that dysregulated autoimmune response in type 1 diabetes may contribute to endothelial dysfunction by increasing oxidative stress through activation of NADPH oxidase [42]. It remains unclear whether glucose fluctuations are involved in endothelial dysfunction in patients with type 1 diabetes due to the paucity of clinical evidence.

4. Recommended Pharmacotherapies from a Perspective of Endothelial Dysfunction in Diabetes Mellitus

Selecting an appropriate intervention that will effectively improve or augment endothelial function is clinically important for preventing cardiovascular events in diabetic patients. Considering that endothelial function may be impaired by chronic hyperglycemia, acute glycemic variability, and insulin resistance through increased oxidative stress, behavior modification and pharmacotherapies aimed at reducing blood glucose levels without hypoglycemia, reducing glucose variability, and ameliorating insulin sensitivity are expected to improve endothelial function.

4.1. Insulin Treatment

Intensive glucose control with insulin has been shown to reduce microvascular and macrovascular complications in patients with type 1 diabetes [43]. Insulin therapy may be beneficial for endothelial function in patients with type 1 diabetes who have a healthy energy balance without insulin resistance since there is little concern about selective insulin resistance. In contrast, the effect of insulin treatment on endothelial function may depend on the achieved level of metabolic control in patients with type 2 diabetes [44]. In obese or overweight patients with type 2 diabetes who have insulin resistance due to overnutrition and a positive energy balance, endothelial function is potentially impaired by high-dose insulin therapy due to selective insulin resistance.

4.2. Hypoglycemic Drugs

Sulfonylureas are insulin secretagogues. Therefore, sulfonylureas, as well as high-dose insulin therapy, could have an adverse effect on endothelial function in overweight or obese diabetics because of the selective insulin resistance, potentially resulting in endothelial dysfunction (Table 1).
Hypoglycemic drugs that reduce glucose fluctuations by decreasing postprandial hyperglycemia are expected to ameliorate endothelial function through decreasing oxidative stress. Glinides, α-glucosidase inhibitors, and dipeptidyl peptidase 4 (DPP-4) inhibitors are hypoglycemic drugs that decrease postprandial hyperglycemia. Indeed, clinical studies have shown that those hypoglycemic drugs improve postprandial endothelial function in diabetic patients [45,46,47,48]. However, there are conflicting reports regarding the effects of glinides, α-glucosidase inhibitors, and DPP-4 inhibitors on postprandial endothelial function; some studies have shown that glinides, DPP-4 inhibitors, and α-glucosidase inhibitors have no beneficial effects on endothelial function [45,49,50]. As for DPP-4 inhibitors and glinides, increased secretion of endogenous insulin through their pharmacological actions is possibly associated with a lack of a beneficial effect of DPP-4 inhibitors or glinides on endothelial function because of the selective insulin resistance in overweight or obese patients with diabetes mellitus.
Glucagon-like peptide 1 receptor (GLP-1R) agonists improve the control of postprandial blood glucose levels. Therefore, GLP-1R agonists are expected to improve endothelial function through reducing glucose fluctuations in patients with diabetes mellitus. Indeed, GLP-1R agonists have been shown to augment endothelial function in patients with diabetes mellitus [51,52,53,54,55]. However, augmentation of postprandial endothelial function by GLP-1R agonists was independent of postprandial changes in blood glucose levels or insulin sensitivity but was associated with postprandial reduction in serum levels of triglycerides [51,52,54]. It has also been shown that eNOS activation in endothelial cells through GLP1-R and AMP-activated protein kinase (AMPK) activation is involved in the augmentation of endothelial function by GLP-1R agonists [53].
Considering that there is a significant association between endothelial dysfunction and insulin resistance, insulin sensitizers are anticipated to ameliorate endothelial function through restoring PI3-kinase/Akt/eNOS pathway and increasing NO production. Indeed, clinical studies have shown that thiazolidinediones, which are insulin sensitizers, improve endothelium-dependent vasodilation [56,57].
Metformin, which is an insulin sensitizer, has been shown to ameliorate endothelial function with a significant association between endothelial function and insulin resistance following treatment in patients with type 2 diabetes mellitus [58]. In addition, metformin has been shown to improve endothelial function in patients without diabetes who have insulin resistance [59]. These findings suggest that metformin improves endothelial function by ameliorating insulin resistance. On the other hand, other clinical studies have shown that metformin improves endothelial dysfunction irrespective of insulin sensitivity or body weight, suggesting that metformin improves endothelial function through multiple mechanisms, some of which are independent of insulin resistance [60,61]. Preclinical studies have shown that metformin improves endothelial function by eNOS phosphorylation through AMPK activation [62,63], sirtuin-1 activation [64,65], and promotion of antioxidation [66].
Sodium-glucose cotransporter 2 (SGLT2) inhibitors lower blood glucose levels by increasing glucose excretion into urine through inhibition of renal glucose reabsorption [67]. The glucose-lowering effect of SGLT2 inhibitors is independent of insulin. Therefore, there is little concern that treatment with SGLT2 inhibitors in overweight or obese diabetics with insulin resistance would further deteriorate endothelial function through the mechanism of selective insulin resistance in endothelial cells. SGLT2 inhibitors have been shown to reduce postprandial glucose levels and decrease overall glucose variability in patients with diabetes mellitus [68,69]. In addition, SGLT2 inhibitors have been shown to reduce insulin resistance and ameliorate peripheral insulin sensitivity [70,71,72]. SGLT2 inhibitors have other metabolic actions including reductions in plasma lipid levels, blood pressure, and body weight. Therefore, SGLT2 inhibitors are expected to improve or augment endothelial function by decreasing oxidative stress through lowering glucose levels in an insulin-independent manner, reducing acute glucose fluctuations, improving insulin sensitivity, and improving other metabolic parameters. Indeed, an experimental study demonstrated that urinary excretion of 8-oxo-2′-deoxyguanosine, an oxidative stress marker, was decreased and endothelial function was ameliorated by treatment with an SGLT2 inhibitor in mice [73]. In addition, clinical studies have shown that SGLT2 inhibitors improve endothelial function in patients with diabetes mellitus [74,75,76,77,78].

4.3. Other Treatment

Dyslipidemia and hypertension are significantly associated with endothelial dysfunction and are highly prevalent in patients with type 2 diabetes [79,80]. Therefore, a multiple-risk-factor intervention approach should be performed to improve endothelial function and prevent future cardiovascular events. The Steno-2 Study demonstrated that the risk of microvascular complications and cardiovascular events was significantly reduced by an intensified, target-driven, multifactorial intervention involving a combination of focused behavior modification and medications aimed at concomitant cardiovascular risk factors than by a conventional strategy [81,82]. In the treatment of other modifiable risk factors in diabetic patients, it is recommended to select an intervention that ameliorates endothelial function, such as administration of statin [83], administration of blockers of the renin-angiotensin system [84,85,86], and behavior modifications such as body weight reduction, aerobic exercise, and smoking cessation [87,88,89,90].
Since oxidative stress has been considered as a major cause of endothelial dysfunction in patients with diabetes, an intervention targeting direct reduction in oxidative stress is attractive and expected to improve endothelial function and cardiovascular outcomes in patients with diabetes. Indeed, endothelial function is augmented by concomitant intra-arterial infusion of vitamin C in patients with diabetes [91,92]. However, oral administration of antioxidants, including vitamin C, vitamin E and N-acetylcysteine, has failed to demonstrate a protective effect of antioxidants against diabetic vascular complications in patients with diabetes [93,94,95,96]. Although the precise reasons for the ineffectiveness of oral administration of antioxidants remain unclear, ineffectiveness may be due to the lack of pharmacokinetic evaluation; plasma levels of the supplemented antioxidants were not monitored and the drug range for safety and efficacy was not determined [12]. Therefore, administration of antioxidants for the prevention of diabetic vascular complications is not clinically recommended in patients with diabetes.

5. Conclusions

Endothelial dysfunction is a therapeutic target in patients with diabetes mellitus. Oxidative stress induced by chronic hyperglycemia, acute glucose fluctuations, and decreased NO production by selective insulin resistance in endothelial cells may be associated with endothelial dysfunction in patients with diabetes. In addition to behavior modifications including body weight reduction, aerobic exercise, and smoking cessation, hypoglycemic drugs that reduce acute glucose fluctuations, such as glinides, α-glucosidase inhibitors and DPP-4 inhibitors, and hypoglycemic drugs that ameliorate insulin sensitivity, such as thiazolidinediones and metformin, are expected to improve endothelial function in patients with diabetes mellitus. Preclinical studies have indicated the possibility that GLP1-R agonists, metformin, and SGLT2 inhibitors improve endothelial function by multiple mechanisms, some of which are independent of glucose control or insulin signaling, such as eNOS phosphorylation through AMPK and sirtuin-1 activation. Selecting appropriate hypoglycemic drugs that will improve or augment endothelial function may be clinically important to prevent diabetic vascular complications for better prognosis in diabetic patients. In the treatment of other modifiable risk factors, including hypertension and dyslipidemia, in patients with diabetes, it is recommended to select an appropriate intervention that will improve endothelial function, such as administration of statins and renin-angiotensin system blockers. Although an intervention targeting direct reduction in oxidative stress by antioxidants is attractive, clinical studies in which the efficacy of oral administration of antioxidants against diabetic vascular complications was investigated have yielded disappointing results. Further studies are needed to develop therapeutic strategies for improving endothelial function and cardiovascular outcomes by decreasing oxidative stress in patients with diabetes.

Author Contributions

Conceptualization, T.M. and Y.H.; writing—original draft preparation, T.M.; writing—review and editing, Y.H.; funding acquisition, T.M. and Y.H. Both authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by Grant-in-Aid for Scientific Research from the Ministry of Education, Culture, Sports, Science and Technology, grant number18590815 and 21590898 to Y.H. and 16K19408 and 19K17565 to T.M. and Grant in Aid of Japanese Arteriosclerosis Prevention Fund (to Y.H.).

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

NO = nitric oxide, ET-1 = endothelin-1, ROS = reactive oxygen species, NADPH = nicotinamide-adenine dinucleotide phosphate, eNOS = endothelial nitric oxide synthase, BH4 = tetrahydrobiopterin, TCA = tricarboxylic acid, NADH = nicotinamide adenine dinucleotide, FADH2 = 1,5-dihydro-flavin adenine dinucleotide, PKC = protein kinase C, AGE = advanced glycation end products, HbA1c = glycated hemoglobin, IRS = insulin receptor substrates, PI3-kinase = phosphoinositide 3-kinase, MAPK = mitogen-activated protein kinase, ERK = extracellular signal-regulated kinase, DPP-4 = dipeptidyl peptidase 4, GLP-1R = glucagon-like peptide 1 receptor, AMPK = AMP-activated protein kinase, SGLT2 = sodium-glucose cotransporter 2.

References

  1. Smolen, J.S.; Burmester, G.R.; Combeet, B. NCD Risk Factor Collaboration. Worldwide trends in diabetes since 1980: A pooled analysis of 751 population-based studies with 4.4 million participants. Lancet 2016, 387, 1513–1530. [Google Scholar]
  2. Einarson, T.R.; Acs, A.; Ludwig, C.; Panton, U.H. Prevalence of cardiovascular disease in type 2 diabetes: A systematic literature review of scientific evidence from across the world in 2007–2017. Cardiovasc. Diabetol. 2018, 17, 83. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Higashi, Y.; Noma, K.; Yoshizumi, M.; Kihara, Y. Endothelial function and oxidative stress in cardiovascular diseases. Circ. J. Off. J. Jpn. Circ. Soc. 2009, 73, 411–418. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Vanhoutte, P.M.; Shimokawa, H.; Feletou, M.; Tang, E.H. Endothelial dysfunction and vascular disease—A 30th anniversary update. Acta Physiol. 2017, 219, 22–96. [Google Scholar] [CrossRef] [PubMed]
  5. Gimbrone, M.A., Jr.; Garcia-Cardena, G. Endothelial Cell Dysfunction and the Pathobiology of Atherosclerosis. Circ. Res. 2016, 118, 620–636. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. Williams, S.B.; Cusco, J.A.; Roddy, M.A.; Johnstone, M.T.; Creager, M.A. Impaired nitric oxide-mediated vasodilation in patients with non-insulin-dependent diabetes mellitus. J. Am. Coll. Cardiol. 1996, 27, 567–574. [Google Scholar] [CrossRef] [Green Version]
  7. Henry, R.M.; Ferreira, I.; Kostense, P.J.; Dekker, J.M.; Nijpels, G.; Heine, R.J.; Kamp, O.; Bouter, L.M.; Stehouwer, C.D. Type 2 diabetes is associated with impaired endothelium-dependent, flow-mediated dilation, but impaired glucose metabolism is not; The Hoorn Study. Atherosclerosis 2004, 174, 49–56. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  8. Maruhashi, T.; Soga, J.; Fujimura, N.; Idei, N.; Mikami, S.; Iwamoto, Y.; Kajikawa, M.; Matsumoto, T.; Hidaka, T.; Kihara, Y.; et al. Relationship between flow-mediated vasodilation and cardiovascular risk factors in a large community-based study. Heart 2013, 99, 1837–1842. [Google Scholar] [CrossRef] [PubMed]
  9. Shi, Y.; Vanhoutte, P.M. Macro- and microvascular endothelial dysfunction in diabetes. J. Diabetes 2017, 9, 434–449. [Google Scholar] [CrossRef] [Green Version]
  10. Vane, J.R.; Anggard, E.E.; Botting, R.M. Regulatory functions of the vascular endothelium. N. Engl. J. Med. 1990, 323, 27–36. [Google Scholar]
  11. Yuan, T.; Yang, T.; Chen, H.; Fu, D.; Hu, Y.; Wang, J.; Yuan, Q.; Yu, H.; Xu, W.; Xie, X. New insights into oxidative stress and inflammation during diabetes mellitus-accelerated atherosclerosis. Redox Biol. 2019, 20, 247–260. [Google Scholar] [CrossRef]
  12. Iacobini, C.; Vitale, M.; Pesce, C.; Pugliese, G.; Menini, S. Diabetic Complications and Oxidative Stress: A 20-Year Voyage Back in Time and Back to the Future. Antioxidants 2021, 10, 727. [Google Scholar] [CrossRef] [PubMed]
  13. Shah, M.S.; Brownlee, M. Molecular and Cellular Mechanisms of Cardiovascular Disorders in Diabetes. Circ. Res. 2016, 118, 1808–1829. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. McVeigh, G.E.; Brennan, G.M.; Johnston, G.D.; McDermott, B.J.; McGrath, L.T.; Henry, W.R.; Andrews, J.W.; Hayes, J.R. Impaired endothelium-dependent and independent vasodilation in patients with type 2 (non-insulin-dependent) diabetes mellitus. Diabetologia 1992, 35, 771–776. [Google Scholar] [PubMed]
  15. Johnstone, M.T.; Creager, S.J.; Scales, K.M.; Cusco, J.A.; Lee, B.K.; Creager, M.A. Impaired endothelium-dependent vasodilation in patients with insulin-dependent diabetes mellitus. Circulation 1993, 88, 2510–2516. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Sies, H.; Jones, D.P. Reactive oxygen species (ROS) as pleiotropic physiological signalling agents. Nat. Rev. Mol. Cell Biol. 2020, 21, 363–383. [Google Scholar] [CrossRef]
  17. Forstermann, U.; Xia, N.; Li, H. Roles of Vascular Oxidative Stress and Nitric Oxide in the Pathogenesis of Atherosclerosis. Circ. Res. 2017, 120, 713–735. [Google Scholar] [CrossRef] [PubMed]
  18. Bartesaghi, S.; Radi, R. Fundamentals on the biochemistry of peroxynitrite and protein tyrosine nitration. Redox Biol. 2018, 14, 618–625. [Google Scholar] [CrossRef]
  19. Forstermann, U.; Sessa, W.C. Nitric oxide synthases: Regulation and function. Eur. Heart J. 2012, 33, 829–837, 837a–837d. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  20. Murphy, M.P. How mitochondria produce reactive oxygen species. Biochem. J. 2009, 417, 1–13. [Google Scholar] [CrossRef] [Green Version]
  21. Brownlee, M. Biochemistry and molecular cell biology of diabetic complications. Nature 2001, 414, 813–820. [Google Scholar]
  22. Du, X.L.; Edelstein, D.; Dimmeler, S.; Ju, Q.; Sui, C.; Brownlee, M. Hyperglycemia inhibits endothelial nitric oxide synthase activity by posttranslational modification at the Akt site. J. Clin. Investig. 2001, 108, 1341–1348. [Google Scholar] [CrossRef]
  23. Ceriello, A. The emerging role of post-prandial hyperglycaemic spikes in the pathogenesis of diabetic complications. Diabet. Med. 1998, 15, 188–193. [Google Scholar] [CrossRef]
  24. Risso, A.; Mercuri, F.; Quagliaro, L.; Damante, G.; Ceriello, A. Intermittent high glucose enhances apoptosis in human umbilical vein endothelial cells in culture. Am. J. Physiol. Endocrinol. Metab. 2001, 281, E924–E930. [Google Scholar] [CrossRef]
  25. Quagliaro, L.; Piconi, L.; Assaloni, R.; Martinelli, L.; Motz, E.; Ceriello, A. Intermittent high glucose enhances apoptosis related to oxidative stress in human umbilical vein endothelial cells: The role of protein kinase C and NAD(P)H-oxidase activation. Diabetes 2003, 52, 2795–2804. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Monnier, L.; Mas, E.; Ginet, C.; Michel, F.; Villon, L.; Cristol, J.P.; Colette, C. Activation of oxidative stress by acute glucose fluctuations compared with sustained chronic hyperglycemia in patients with type 2 diabetes. JAMA 2006, 295, 1681–1687. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Torimoto, K.; Okada, Y.; Mori, H.; Tanaka, Y. Relationship between fluctuations in glucose levels measured by continuous glucose monitoring and vascular endothelial dysfunction in type 2 diabetes mellitus. Cardiovasc. Diabetol. 2013, 12, 1. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  28. Group, A.C.; Patel, A.; MacMahon, S.; Chalmers, J.; Neal, B.; Billot, L.; Woodward, M.; Marre, M.; Cooper, M.; Glasziou, P.; et al. Intensive blood glucose control and vascular outcomes in patients with type 2 diabetes. N. Engl. J. Med. 2008, 358, 2560–2572. [Google Scholar]
  29. Group, A.S.; Gerstein, H.C.; Miller, M.E.; Genuth, S.; Ismail-Beigi, F.; Buse, J.B.; Goff, D.C., Jr.; Probstfield, J.L.; Cushman, W.C.; Ginsberg, H.N.; et al. Long-term effects of intensive glucose lowering on cardiovascular outcomes. N. Engl. J. Med. 2011, 364, 818–828. [Google Scholar] [CrossRef] [Green Version]
  30. Duckworth, W.; Abraira, C.; Moritz, T.; Reda, D.; Emanuele, N.; Reaven, P.D.; Zieve, F.J.; Marks, J.; Davis, S.N.; Hayward, R.; et al. Glucose control and vascular complications in veterans with type 2 diabetes. N. Engl. J. Med. 2009, 360, 129–139. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  31. Zeng, G.; Quon, M.J. Insulin-stimulated production of nitric oxide is inhibited by wortmannin. Direct measurement in vascular endothelial cells. J. Clin. Investig. 1996, 98, 894–898. [Google Scholar] [CrossRef] [Green Version]
  32. Zeng, G.; Nystrom, F.H.; Ravichandran, L.V.; Cong, L.N.; Kirby, M.; Mostowski, H.; Quon, M.J. Roles for insulin receptor, PI3-kinase, and Akt in insulin-signaling pathways related to production of nitric oxide in human vascular endothelial cells. Circulation 2000, 101, 1539–1545. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Gogg, S.; Smith, U.; Jansson, P.A. Increased MAPK activation and impaired insulin signaling in subcutaneous microvascular endothelial cells in type 2 diabetes: The role of endothelin-1. Diabetes 2009, 58, 2238–2245. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Kubota, T.; Kubota, N.; Kadowaki, T. The role of endothelial insulin signaling in the regulation of glucose metabolism. Rev. Endocr. Metab. Disord. 2013, 14, 207–216. [Google Scholar] [CrossRef]
  35. Clarkson, P.; Celermajer, D.S.; Donald, A.E.; Sampson, M.; Sorensen, K.E.; Adams, M.; Yue, D.K.; Betteridge, D.J.; Deanfield, J.E. Impaired vascular reactivity in insulin-dependent diabetes mellitus is related to disease duration and low density lipoprotein cholesterol levels. J. Am. Coll. Cardiol. 1996, 28, 573–579. [Google Scholar] [CrossRef] [Green Version]
  36. Varvarovska, J.; Racek, J.; Stetina, R.; Sykora, J.; Pomahacova, R.; Rusavy, Z.; Lacigova, S.; Trefil, L.; Siala, K.; Stozicky, F. Aspects of oxidative stress in children with type 1 diabetes mellitus. Biomed. Pharmacother. Biomed. Pharmacother. 2004, 58, 539–545. [Google Scholar] [CrossRef]
  37. Varvarovska, J.; Racek, J.; Stozicky, F.; Soucek, J.; Trefil, L.; Pomahacova, R. Parameters of oxidative stress in children with Type 1 diabetes mellitus and their relatives. J. Diabetes Complicat. 2003, 17, 7–10. [Google Scholar] [CrossRef]
  38. Dominguez, C.; Ruiz, E.; Gussinye, M.; Carrascosa, A. Oxidative stress at onset and in early stages of type 1 diabetes in children and adolescents. Diabetes Care 1998, 21, 1736–1742. [Google Scholar] [CrossRef]
  39. Ladeia, A.M.; Ladeia-Frota, C.; Pinho, L.; Stefanelli, E.; Adan, L. Endothelial dysfunction is correlated with microalbuminuria in children with short-duration type 1 diabetes. Diabetes Care 2005, 28, 2048–2050. [Google Scholar] [CrossRef] [Green Version]
  40. Khan, F.; Elhadd, T.A.; Greene, S.A.; Belch, J.J. Impaired skin microvascular function in children, adolescents, and young adults with type 1 diabetes. Diabetes Care 2000, 23, 215–220. [Google Scholar] [CrossRef] [Green Version]
  41. Ce, G.V.; Rohde, L.E.; da Silva, A.M.; Punales, M.K.; de Castro, A.C.; Bertoluci, M.C. Endothelial dysfunction is related to poor glycemic control in adolescents with type 1 diabetes under 5 years of disease: Evidence of metabolic memory. J. Clin. Endocrinol. Metab. 2011, 96, 1493–1499. [Google Scholar] [CrossRef] [Green Version]
  42. Zimmerman, M.A.; Flores, S.C. Autoimmune-mediated oxidative stress and endothelial dysfunction: Implications of accelerated vascular injury in type I diabetes. J. Surg. Res. 2009, 155, 173–178. [Google Scholar] [CrossRef] [PubMed]
  43. The Diabetes Control and Complications Trial/Epidemiology of Diabetes Interventions and Complications (DCCT/EDIC) Study Research Group. Intensive diabetes treatment and cardiovascular disease in patients with type 1 diabetes. N. Engl. J. Med. 2005, 353, 2643–2653. [Google Scholar] [PubMed]
  44. Nolan, C.J.; Ruderman, N.B.; Kahn, S.E.; Pedersen, O.; Prentki, M. Insulin resistance as a physiological defense against metabolic stress: Implications for the management of subsets of type 2 diabetes. Diabetes 2015, 64, 673–686. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Kato, T.; Inoue, T.; Node, K. Postprandial endothelial dysfunction in subjects with new-onset type 2 diabetes: An acarbose and nateglinide comparative study. Cardiovasc. Diabetol. 2010, 9, 12. [Google Scholar] [CrossRef] [Green Version]
  46. Nakamura, K.; Oe, H.; Kihara, H.; Shimada, K.; Fukuda, S.; Watanabe, K.; Takagi, T.; Yunoki, K.; Miyoshi, T.; Hirata, K.; et al. DPP-4 inhibitor and alpha-glucosidase inhibitor equally improve endothelial function in patients with type 2 diabetes: EDGE study. Cardiovasc. Diabetol. 2014, 13, 110. [Google Scholar] [CrossRef] [Green Version]
  47. Shimabukuro, M.; Higa, N.; Chinen, I.; Yamakawa, K.; Takasu, N. Effects of a single administration of acarbose on postprandial glucose excursion and endothelial dysfunction in type 2 diabetic patients: A randomized crossover study. J. Clin. Endocrinol. Metab. 2006, 91, 837–842. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  48. Matsubara, J.; Sugiyama, S.; Akiyama, E.; Iwashita, S.; Kurokawa, H.; Ohba, K.; Maeda, H.; Fujisue, K.; Yamamoto, E.; Kaikita, K.; et al. Dipeptidyl peptidase-4 inhibitor, sitagliptin, improves endothelial dysfunction in association with its anti-inflammatory effects in patients with coronary artery disease and uncontrolled diabetes. Circ. J. Off. J. Jpn. Circ. Soc. 2013, 77, 1337–1344. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  49. Ayaori, M.; Iwakami, N.; Uto-Kondo, H.; Sato, H.; Sasaki, M.; Komatsu, T.; Iizuka, M.; Takiguchi, S.; Yakushiji, E.; Nakaya, K.; et al. Dipeptidyl peptidase-4 inhibitors attenuate endothelial function as evaluated by flow-mediated vasodilatation in type 2 diabetic patients. J. Am. Heart Assoc. 2013, 2, e003277. [Google Scholar] [CrossRef] [Green Version]
  50. Maruhashi, T.; Higashi, Y.; Kihara, Y.; Yamada, H.; Sata, M.; Ueda, S.; Odawara, M.; Terauchi, Y.; Dai, K.; Ohno, J.; et al. Long-term effect of sitagliptin on endothelial function in type 2 diabetes: A sub-analysis of the PROLOGUE study. Cardiovasc. Diabetol. 2016, 15, 134. [Google Scholar] [CrossRef] [Green Version]
  51. Nystrom, T.; Gutniak, M.K.; Zhang, Q.; Zhang, F.; Holst, J.J.; Ahren, B.; Sjoholm, A. Effects of glucagon-like peptide-1 on endothelial function in type 2 diabetes patients with stable coronary artery disease. Am. J. Physiol. Endocrinol. Metab. 2004, 287, E1209–E1215. [Google Scholar] [CrossRef] [PubMed]
  52. Koska, J.; Schwartz, E.A.; Mullin, M.P.; Schwenke, D.C.; Reaven, P.D. Improvement of postprandial endothelial function after a single dose of exenatide in individuals with impaired glucose tolerance and recent-onset type 2 diabetes. Diabetes Care 2010, 33, 1028–1030. [Google Scholar] [CrossRef] [Green Version]
  53. Koska, J.; Sands, M.; Burciu, C.; D’Souza, K.M.; Raravikar, K.; Liu, J.; Truran, S.; Franco, D.A.; Schwartz, E.A.; Schwenke, D.C.; et al. Exenatide Protects Against Glucose- and Lipid-Induced Endothelial Dysfunction: Evidence for Direct Vasodilation Effect of GLP-1 Receptor Agonists in Humans. Diabetes 2015, 64, 2624–2635. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Torimoto, K.; Okada, Y.; Mori, H.; Otsuka, T.; Kawaguchi, M.; Matsuda, M.; Kuno, F.; Sugai, K.; Sonoda, S.; Hajime, M.; et al. Effects of exenatide on postprandial vascular endothelial dysfunction in type 2 diabetes mellitus. Cardiovasc. Diabetol. 2015, 14, 25. [Google Scholar] [CrossRef] [Green Version]
  55. Tuttolomondo, A.; Cirrincione, A.; Casuccio, A.; Del Cuore, A.; Daidone, M.; Di Chiara, T.; Di Raimondo, D.; Corte, V.D.; Maida, C.; Simonetta, I.; et al. Efficacy of dulaglutide on vascular health indexes in subjects with type 2 diabetes: A randomized trial. Cardiovasc. Diabetol. 2021, 20, 1. [Google Scholar] [CrossRef] [PubMed]
  56. Fujishima, S.; Ohya, Y.; Nakamura, Y.; Onaka, U.; Abe, I.; Fujishima, M. Troglitazone, an insulin sensitizer, increases forearm blood flow in humans. Am. J. Hypertens. 1998, 11, 1134–1137. [Google Scholar] [CrossRef]
  57. Hidaka, T.; Nakagawa, K.; Goto, C.; Soga, J.; Fujii, Y.; Hata, T.; Idei, N.; Fujimura, N.; Chayama, K.; Kihara, Y.; et al. Pioglitazone improves endothelium-dependent vasodilation in hypertensive patients with impaired glucose tolerance in part through a decrease in oxidative stress. Atherosclerosis 2010, 210, 521–524. [Google Scholar] [CrossRef] [PubMed]
  58. Mather, K.J.; Verma, S.; Anderson, T.J. Improved endothelial function with metformin in type 2 diabetes mellitus. J. Am. Coll. Cardiol. 2001, 37, 1344–1350. [Google Scholar] [CrossRef] [Green Version]
  59. Vitale, C.; Mercuro, G.; Cornoldi, A.; Fini, M.; Volterrani, M.; Rosano, G.M. Metformin improves endothelial function in patients with metabolic syndrome. J. Intern. Med. 2005, 258, 250–256. [Google Scholar] [CrossRef]
  60. Naka, K.K.; Papathanassiou, K.; Bechlioulis, A.; Pappas, K.; Kazakos, N.; Kanioglou, C.; Kostoula, A.; Vezyraki, P.; Makriyiannis, D.; Tsatsoulis, A.; et al. Effects of pioglitazone and metformin on vascular endothelial function in patients with type 2 diabetes treated with sulfonylureas. Diabetes Vasc. Dis. Res. 2012, 9, 52–58. [Google Scholar] [CrossRef] [Green Version]
  61. Pitocco, D.; Zaccardi, F.; Tarzia, P.; Milo, M.; Scavone, G.; Rizzo, P.; Pagliaccia, F.; Nerla, R.; Di Franco, A.; Manto, A.; et al. Metformin improves endothelial function in type 1 diabetic subjects: A pilot, placebo-controlled randomized study. Diabetes Obes. Metab. 2013, 15, 427–431. [Google Scholar] [CrossRef]
  62. Chen, Z.P.; Mitchelhill, K.I.; Michell, B.J.; Stapleton, D.; Rodriguez-Crespo, I.; Witters, L.A.; Power, D.A.; Ortiz de Montellano, P.R.; Kemp, B.E. AMP-activated protein kinase phosphorylation of endothelial NO synthase. FEBS Lett. 1999, 443, 285–289. [Google Scholar] [CrossRef] [Green Version]
  63. Davis, B.J.; Xie, Z.; Viollet, B.; Zou, M.H. Activation of the AMP-activated kinase by antidiabetes drug metformin stimulates nitric oxide synthesis in vivo by promoting the association of heat shock protein 90 and endothelial nitric oxide synthase. Diabetes 2006, 55, 496–505. [Google Scholar] [CrossRef] [Green Version]
  64. Mattagajasingh, I.; Kim, C.S.; Naqvi, A.; Yamamori, T.; Hoffman, T.A.; Jung, S.B.; DeRicco, J.; Kasuno, K.; Irani, K. SIRT1 promotes endothelium-dependent vascular relaxation by activating endothelial nitric oxide synthase. Proc. Natl. Acad. Sci. USA 2007, 104, 14855–14860. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Arunachalam, G.; Samuel, S.M.; Marei, I.; Ding, H.; Triggle, C.R. Metformin modulates hyperglycaemia-induced endothelial senescence and apoptosis through SIRT1. Br. J. Pharmacol. 2014, 171, 523–535. [Google Scholar] [CrossRef] [PubMed]
  66. Salvatore, T.; Pafundi, P.C.; Galiero, R.; Rinaldi, L.; Caturano, A.; Vetrano, E.; Aprea, C.; Albanese, G.; Di Martino, A.; Ricozzi, C.; et al. Can Metformin Exert as an Active Drug on Endothelial Dysfunction in Diabetic Subjects? Biomedicines 2020, 9, 3. [Google Scholar] [CrossRef]
  67. Gallo, L.A.; Wright, E.M.; Vallon, V. Probing SGLT2 as a therapeutic target for diabetes: Basic physiology and consequences. Diabetes Vasc. Dis. Res. 2015, 12, 78–89. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  68. Rodbard, H.W.; Peters, A.L.; Slee, A.; Cao, A.; Traina, S.B.; Alba, M. The Effect of Canagliflozin, a Sodium Glucose Cotransporter 2 Inhibitor, on Glycemic End Points Assessed by Continuous Glucose Monitoring and Patient-Reported Outcomes Among People With Type 1 Diabetes. Diabetes Care 2017, 40, 171–180. [Google Scholar] [CrossRef] [Green Version]
  69. Henry, R.R.; Strange, P.; Zhou, R.; Pettus, J.; Shi, L.; Zhuplatov, S.B.; Mansfield, T.; Klein, D.; Katz, A. Effects of Dapagliflozin on 24-Hour Glycemic Control in Patients with Type 2 Diabetes: A Randomized Controlled Trial. Diabetes Technol. Ther. 2018, 20, 715–724. [Google Scholar] [CrossRef] [Green Version]
  70. Merovci, A.; Solis-Herrera, C.; Daniele, G.; Eldor, R.; Fiorentino, T.V.; Tripathy, D.; Xiong, J.; Perez, Z.; Norton, L.; Abdul-Ghani, M.A.; et al. Dapagliflozin improves muscle insulin sensitivity but enhances endogenous glucose production. J. Clin. Investig. 2014, 124, 509–514. [Google Scholar] [CrossRef]
  71. Ferrannini, E.; Muscelli, E.; Frascerra, S.; Baldi, S.; Mari, A.; Heise, T.; Broedl, U.C.; Woerle, H.J. Metabolic response to sodium-glucose cotransporter 2 inhibition in type 2 diabetic patients. J. Clin. Investig. 2014, 124, 499–508. [Google Scholar] [CrossRef] [Green Version]
  72. Obata, A.; Kubota, N.; Kubota, T.; Iwamoto, M.; Sato, H.; Sakurai, Y.; Takamoto, I.; Katsuyama, H.; Suzuki, Y.; Fukazawa, M.; et al. Tofogliflozin Improves Insulin Resistance in Skeletal Muscle and Accelerates Lipolysis in Adipose Tissue in Male Mice. Endocrinology 2016, 157, 1029–1042. [Google Scholar] [CrossRef] [PubMed]
  73. Rahadian, A.; Fukuda, D.; Salim, H.M.; Yagi, S.; Kusunose, K.; Yamada, H.; Soeki, T.; Sata, M. Canagliflozin Prevents Diabetes-Induced Vascular Dysfunction in ApoE-Deficient Mice. J. Atheroscler. Thromb. 2020, 27, 1141–1151. [Google Scholar] [CrossRef] [Green Version]
  74. Sugiyama, S.; Jinnouchi, H.; Kurinami, N.; Hieshima, K.; Yoshida, A.; Jinnouchi, K.; Nishimura, H.; Suzuki, T.; Miyamoto, F.; Kajiwara, K.; et al. The SGLT2 Inhibitor Dapagliflozin Significantly Improves the Peripheral Microvascular Endothelial Function in Patients with Uncontrolled Type 2 Diabetes Mellitus. Intern. Med. 2018, 57, 2147–2156. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Sakai, T.; Miura, S. Effects of Sodium-Glucose Cotransporter 2 Inhibitor on Vascular Endothelial and Diastolic Function in Heart Failure With Preserved Ejection Fraction- Novel Prospective Cohort Study. Circ. Rep. 2019, 1, 286–295. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. Tochiya, M.; Makino, H.; Tamanaha, T.; Matsuo, M.; Hishida, A.; Koezuka, R.; Ohata, Y.; Tomita, T.; Son, C.; Miyamoto, Y.; et al. Effect of tofogliflozin on cardiac and vascular endothelial function in patients with type 2 diabetes and heart diseases: A pilot study. J. Diabetes Investig. 2020, 11, 400–404. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  77. Sawada, T.; Uzu, K.; Hashimoto, N.; Onishi, T.; Takaya, T.; Shimane, A.; Taniguchi, Y.; Yasaka, Y.; Ohara, T.; Kawai, H. Empagliflozin’s Ameliorating Effect on Plasma Triglycerides: Association with Endothelial Function Recovery in Diabetic Patients with Coronary Artery Disease. J. Atheroscler. Thromb. 2020, 27, 644–656. [Google Scholar] [CrossRef] [Green Version]
  78. Shigiyama, F.; Kumashiro, N.; Miyagi, M.; Ikehara, K.; Kanda, E.; Uchino, H.; Hirose, T. Effectiveness of dapagliflozin on vascular endothelial function and glycemic control in patients with early-stage type 2 diabetes mellitus: DEFENCE study. Cardiovasc. Diabetol. 2017, 16, 84. [Google Scholar] [CrossRef] [Green Version]
  79. Maruhashi, T.; Soga, J.; Fujimura, N.; Idei, N.; Mikami, S.; Iwamoto, Y.; Iwamoto, A.; Kajikawa, M.; Matsumoto, T.; Oda, N.; et al. Endothelial Function Is Impaired in Patients Receiving Antihypertensive Drug Treatment Regardless of Blood Pressure Level: FMD-J Study (Flow-Mediated Dilation Japan). Hypertension 2017, 70, 790–797. [Google Scholar] [CrossRef]
  80. Matsui, S.; Kajikawa, M.; Hida, E.; Maruhashi, T.; Iwamoto, Y.; Iwamoto, A.; Oda, N.; Kishimoto, S.; Hidaka, T.; Kihara, Y.; et al. Optimal Target Level of Low-density Lipoprotein Cholesterol for Vascular Function in Statin Naive Individuals. Sci. Rep. 2017, 7, 8422. [Google Scholar] [CrossRef] [Green Version]
  81. Gaede, P.; Vedel, P.; Larsen, N.; Jensen, G.V.; Parving, H.H.; Pedersen, O. Multifactorial intervention and cardiovascular disease in patients with type 2 diabetes. N. Engl. J. Med. 2003, 348, 383–393. [Google Scholar] [CrossRef] [Green Version]
  82. Gaede, P.; Lund-Andersen, H.; Parving, H.H.; Pedersen, O. Effect of a multifactorial intervention on mortality in type 2 diabetes. N. Engl. J. Med. 2008, 358, 580–591. [Google Scholar] [CrossRef] [Green Version]
  83. Wolfrum, S.; Jensen, K.S.; Liao, J.K. Endothelium-dependent effects of statins. Arterioscler. Thromb. Vasc. Biol. 2003, 23, 729–736. [Google Scholar] [CrossRef] [PubMed]
  84. Higashi, Y.; Sasaki, S.; Nakagawa, K.; Ueda, T.; Yoshimizu, A.; Kurisu, S.; Matsuura, H.; Kajiyama, G.; Oshima, T. A comparison of angiotensin-converting enzyme inhibitors, calcium antagonists, beta-blockers and diuretic agents on reactive hyperemia in patients with essential hypertension: A multicenter study. J. Am. Coll. Cardiol. 2000, 35, 284–291. [Google Scholar] [CrossRef] [Green Version]
  85. Ghiadoni, L.; Virdis, A.; Magagna, A.; Taddei, S.; Salvetti, A. Effect of the angiotensin II type 1 receptor blocker candesartan on endothelial function in patients with essential hypertension. Hypertension 2000, 35, 501–506. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Fujimura, N.; Noma, K.; Hata, T.; Soga, J.; Hidaka, T.; Idei, N.; Fujii, Y.; Mikami, S.; Maruhashi, T.; Iwamoto, Y.; et al. Mineralocorticoid receptor blocker eplerenone improves endothelial function and inhibits Rho-associated kinase activity in patients with hypertension. Clin. Pharmacol. Ther. 2012, 91, 289–297. [Google Scholar] [CrossRef] [PubMed]
  87. Higashi, Y.; Sasaki, S.; Kurisu, S.; Yoshimizu, A.; Sasaki, N.; Matsuura, H.; Kajiyama, G.; Oshima, T. Regular aerobic exercise augments endothelium-dependent vascular relaxation in normotensive as well as hypertensive subjects: Role of endothelium-derived nitric oxide. Circulation 1999, 100, 1194–1202. [Google Scholar] [CrossRef] [Green Version]
  88. Sasaki, S.; Higashi, Y.; Nakagawa, K.; Kimura, M.; Noma, K.; Sasaki, S.; Hara, K.; Matsuura, H.; Goto, C.; Oshima, T.; et al. A low-calorie diet improves endothelium-dependent vasodilation in obese patients with essential hypertension. Am. J. Hypertens. 2002, 15, 302–309. [Google Scholar] [CrossRef]
  89. Goto, C.; Higashi, Y.; Kimura, M.; Noma, K.; Hara, K.; Nakagawa, K.; Kawamura, M.; Chayama, K.; Yoshizumi, M.; Nara, I. Effect of different intensities of exercise on endothelium-dependent vasodilation in humans: Role of endothelium-dependent nitric oxide and oxidative stress. Circulation 2003, 108, 530–535. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  90. Celermajer, D.S.; Sorensen, K.E.; Georgakopoulos, D.; Bull, C.; Thomas, O.; Robinson, J.; Deanfield, J.E. Cigarette smoking is associated with dose-related and potentially reversible impairment of endothelium-dependent dilation in healthy young adults. Circulation 1993, 88, 2149–2155. [Google Scholar] [CrossRef] [Green Version]
  91. Ting, H.H.; Timimi, F.K.; Boles, K.S.; Creager, S.J.; Ganz, P.; Creager, M.A. Vitamin C improves endothelium-dependent vasodilation in patients with non-insulin-dependent diabetes mellitus. J. Clin. Investig. 1996, 97, 22–28. [Google Scholar] [CrossRef] [Green Version]
  92. Timimi, F.K.; Ting, H.H.; Haley, E.A.; Roddy, M.A.; Ganz, P.; Creager, M.A. Vitamin C improves endothelium-dependent vasodilation in patients with insulin-dependent diabetes mellitus. J. Am. Coll. Cardiol. 1998, 31, 552–557. [Google Scholar] [CrossRef] [Green Version]
  93. Lee, D.H.; Folsom, A.R.; Harnack, L.; Halliwell, B.; Jacobs, D.R., Jr. Does supplemental vitamin C increase cardiovascular disease risk in women with diabetes? Am. J. Clin. Nutr. 2004, 80, 1194–1200. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Saklayen, M.G.; Yap, J.; Vallyathan, V. Effect of month-long treatment with oral N-acetylcysteine on the oxidative stress and proteinuria in patients with diabetic nephropathy: A pilot study. J. Investig. Med. Off. Publ. Am. Fed. Clin. Res. 2010, 58, 28–31. [Google Scholar] [CrossRef]
  95. The Vitamin E in Neuroprotection Study (VENUS) Investigators; Hor, C.P.; Fung, W.Y.; Ang, H.A.; Lim, S.C.; Kam, L.Y.; Sim, S.W.; Lim, L.H.; Choon, W.Y.; Wong, J.W.; et al. Efficacy of Oral Mixed Tocotrienols in Diabetic Peripheral Neuropathy: A Randomized Clinical Trial. JAMA Neurol. 2018, 75, 444–452. [Google Scholar] [PubMed]
  96. Lonn, E.; Bosch, J.; Yusuf, S.; Sheridan, P.; Pogue, J.; Arnold, J.M.; Ross, C.; Arnold, A.; Sleight, P.; Probstfield, J.; et al. Effects of long-term vitamin E supplementation on cardiovascular events and cancer: A randomized controlled trial. JAMA 2005, 293, 1338–1347. [Google Scholar] [CrossRef] [Green Version]
Figure 1. Mechanisms of endothelial dysfunction in diabetic patients. Chronic hyperglycemia, acute glucose fluctuations, and insulin resistance are involved in endothelial dysfunction in diabetic patients. ET-1, endothelin-1; ROS, reactive oxygen species; AGE, advanced glycation end products; PI3-kinase, phosphoinositide 3-kinase; MAPK, mitogen-activated protein kinase; ERK, extracellular signal-regulated kinase.
Figure 1. Mechanisms of endothelial dysfunction in diabetic patients. Chronic hyperglycemia, acute glucose fluctuations, and insulin resistance are involved in endothelial dysfunction in diabetic patients. ET-1, endothelin-1; ROS, reactive oxygen species; AGE, advanced glycation end products; PI3-kinase, phosphoinositide 3-kinase; MAPK, mitogen-activated protein kinase; ERK, extracellular signal-regulated kinase.
Antioxidants 10 01306 g001
Table 1. Hypoglycemic drugs and their effects on endothelial function.
Table 1. Hypoglycemic drugs and their effects on endothelial function.
Hypoglycemic DrugsEffects on Endothelial Function
Insulin therapy [44]Endothelial function is potentially impaired due to the selective insulin resistance in patients with insulin resistance.
Sulfonylureas [44]Endothelial function is potentially impaired due to the selective insulin resistance in patients with insulin resistance.
Glinides [45]∙ Endothelial function is potentially improved by reducing glucose fluctuations through decreasing postprandial hyperglycemia.
∙ Endothelial function is potentially impaired due to the selective insulin resistance in patients with insulin resistance.
α-glucosidase inhibitors [45,46,47]Endothelial function is potentially improved by reducing glucose fluctuations through decreasing postprandial hyperglycemia.
DPP-4 inhibitors [46,48,49,50]∙ Endothelial function is potentially improved by reducing glucose fluctuations through decreasing postprandial hyperglycemia.
∙ Endothelial function is potentially impaired due to the selective insulin resistance in patients with insulin resistance.
GLP-1R agonists [51,52,53,54,55]Endothelial function is potentially improved by reducing glucose fluctuations through decreasing postprandial hyperglycemia, by reducing postprandial triglycerides levels, and by activating AMPK.
Thiazolidinediones [56,57]Endothelial function is potentially improved by reducing insulin resistance.
Metformin [58,59,60,61,62,63,64,65,66]Endothelial function is potentially improved by reducing insulin resistance, activating AMPK and sirtuin-1, and promoting antioxidation.
SGLT2 inhibitors [67,68,69,70,71,72,73,74,75,76,77,78]Endothelial function is potentially improved by lowering glucose levels in an insulin-independent manner, reducing acute glucose fluctuations, improving insulin sensitivity, and improving other metabolic parameters.
DPP-4 indicates dipeptidyl peptidase 4; GLP-1R, glucagon-like peptide 1 receptor; SGLT2, sodium-glucose cotransporter 2; AMPK, AMP-activated protein kinase.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Maruhashi, T.; Higashi, Y. Pathophysiological Association between Diabetes Mellitus and Endothelial Dysfunction. Antioxidants 2021, 10, 1306. https://doi.org/10.3390/antiox10081306

AMA Style

Maruhashi T, Higashi Y. Pathophysiological Association between Diabetes Mellitus and Endothelial Dysfunction. Antioxidants. 2021; 10(8):1306. https://doi.org/10.3390/antiox10081306

Chicago/Turabian Style

Maruhashi, Tatsuya, and Yukihito Higashi. 2021. "Pathophysiological Association between Diabetes Mellitus and Endothelial Dysfunction" Antioxidants 10, no. 8: 1306. https://doi.org/10.3390/antiox10081306

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop