Abstract
Diabetes mellitus increases the risk of developing heart failure, and the co-existence of both diseases worsens cardiovascular outcomes, hospitalization, and the progression of heart failure. Despite current advancements on therapeutic strategies to manage hyperglycaemia, the likelihood of developing diabetes-induced heart failure is still significant, especially with the accelerating global prevalence of diabetes and an ageing population. This raises the likelihood of other contributing mechanisms beyond hyperglycaemia in predisposing diabetic patients to cardiovascular disease risk. There has been considerable interest in understanding the alterations in cardiac structure and function in diabetic patients, collectively termed as ‘diabetic cardiomyopathy’. However, the factors that contribute to the development of diabetic cardiomyopathies are not fully understood. This review summarizes the main characteristics of diabetic cardiomyopathies, and the basic mechanisms that contribute to its occurrence. This includes perturbations in insulin resistance, fuel preference, reactive oxygen species generation, inflammation, cell death pathways, neurohormonal mechanisms, advanced glycated end-products accumulation, lipotoxicity, glucotoxicity, and post-translational modifications in the heart of the diabetic. This review also discusses the impact of antihyperglycaemic therapies on the development of heart failure, as well as how current heart failure therapies influence glycaemic control in diabetic patients. We also highlight the current knowledge gaps in understanding how diabetes induces heart failure.
1. Introduction
Cardiovascular disease is the leading cause of death and complications in diabetic patients worldwide,1–6 the prevalence of which is increasing despite therapeutic and pharmacological advances.5,7 Diabetes mellitus, a metabolic disorder characterized by hyperglycaemia resulting from insulin deficiency (type 1) or resistance (type 2).8 is a major independent risk factor in the development of heart failure9–11 and is becoming a global epidemic with increasing prevalence.12–15 There are two main types of diabetes mellitus: type 1 and type 2; gestational diabetes mellitus is also part of this category but is not a major focus of this review. Type 1 diabetes mellitus, also called insulin-dependent diabetes, is an autoimmune disease where pancreatic beta cells are destroyed, and therefore, the body is unable to produce insulin.16 Type 2 diabetes mellitus, also called non-insulin-dependent diabetes, is characterized by a deficit in the function of insulin produced by pancreatic beta cells; this is also referred to as insulin resistance. Type 2 diabetes is the more common form of diabetes, and factors such as age, obesity, diet, and pre-existing hypertension affect its development and its risk of development. Therefore, due to either the elimination of insulin secretion or the reduction in insulin function, blood glucose levels are, as a result, elevated, leading to chronic hyperglycaemia if left untreated. The Framingham Study was one of the first epidemiological studies to show an increased risk of heart failure in patients with diabetes mellitus,10,17,18 with other clinical trials supporting this conclusion (see refs19,20 for reviews). The presence of both diabetes and heart failure in individuals leads to poor cardiovascular outcomes.5,6,21–24 Diabetic patients have higher mortality from coronary artery disease than non-diabetics25 and show a worse prognosis,26,27 which may be associated with increased atherosclerosis,25 and can lead to ischaemic heart failure.28 Heart failure is the major adverse cardiovascular outcome in diabetic patients.18 Poor glycaemic control is associated with increased risk of heart failure in individuals with type 2 diabetes,29 indicated by elevated haemoglobin AIC levels, an index of glycaemic control.30 Therefore, it is critical to mediate and treat cardiovascular conditions in diabetic patients. The prevention and treatment of cardiovascular disease and heart failure remains a considerable challenge in the treatment and management of diabetes mellitus.
Diabetic cardiomyopathy is a condition characterized by ventricular dysfunction and hypertrophy in diabetic patients independent of hypertension, ischaemia, or coronary artery disease.31 The term originated from a Rubler et al. study that identified diabetic patients with congestive heart failure without the aforementioned risk factors or other causes. Non-invasive studies show impaired diastolic and systolic function,32–35 especially with the presence of hypertension. Multiple mechanisms contribute to the development of heart failure in diabetic individuals, including increased inflammation36 and oxidative stress,37,38 changes in cardiac myocardial energy metabolism,39–41 cardiac lipotoxicity,42–45 impaired cardiomyocyte calcium handling, and apoptosis.46,47 Diabetic cardiomyopathy may progress to either heart failure with preserved ejection fraction (HFpEF), where there is diastolic dysfunction,48,49 or heart failure with reduced ejection fraction (HFrEF), where there is systolic dysfunction.11,50,51 Each phenotype can be distinguished by the various mechanisms involved in contributing to it. Hypertrophy,52–54 insulin resistance,55 and lipotoxicity56,57 have been shown to contribute to the development of HFpEF, while oxidative stress,58 fibrosis,59,60 and autoimmunity caused cardiomyocyte cell death61 are involved in contributing to HFrEF. Coronary deposition of advanced glycation end-products is involved in both phenotypes.62 Diastolic dysfunction, which is part of the diagnosis of HFpEF,48,49,63 can precede the development of HFrEF alongside comorbidities of impaired coronary vasculature and endothelial function, and hypertrophy.64,65 Although diastolic dysfunction is mostly predominant in diabetic cardiomyopathy, systolic dysfunction may also occur in later stages of diabetic cardiomyopathy, which can contribute to the development of HFrEF.50,51 Studies have demonstrated a metabolic link between the two heart failure phonotypes whereby abnormal mitochondrial function and oxidative stress can lead from an HFpEF phenotype into an HFrEF phenotype by mediating cardiac hypertrophy, inflammation, fibrosis, and further endothelial cell damage, which has adverse consequences on systolic function, and ultimately more severe manifestations of diabetes and heart failure.52,66–75
The aim of this review is to highlight the changes that occur in diabetic cardiomyopathy, alongside the mechanisms involved its development and progression. The effect of antihyperglycaemic drugs on heart failure risk in diabetic individuals, and heart failure drugs on glycaemic control will also be discussed, as well as novel therapeutic approaches.
2. Structural and functional characteristics of the failing heart in diabetics
The failing heart in the context of diabetes is characterized by multiple alterations including impairments in diastolic and subsequent systolic function,32–34,48 cardiac hypertrophy and fibrosis,76 and impaired coronary microvascular perfusion.77
2.1 Diastolic and systolic dysfunction
Heart failure in diabetes is characterized by cardiac dysfunction, with diastolic dysfunction as a hallmark of the failing myocardium in diabetics.32,78 Echocardiography and Doppler imaging assessments of diastolic dysfunction,79 have shown that left ventricular (LV) dysfunction is manifested in type 2 diabetic patients through altered LV filling,80 abnormal LV relaxation,32,81,82 reduced LV end-diastolic volume,83 and LV chamber stiffness.84 Studies in type 1 diabetics demonstrate abnormalities in LV diastolic filling,33,85,86 lower E to A ratios, prolonged isovolumetric relaxation times,33 and reduction in end-systolic volumes.34 Thus, there is no single parameter to indicate and quantify diastolic dysfunction. Moreover, Attali et al.87 showed in both type 1 and 2 diabetic patients that abnormalities in diastolic function, including increased isovolumetric relaxation time and impaired LV compliance, were not related to additional factors such as age, sex, duration of diabetes, or presence of other complications. Impaired diastolic function has also been shown in type 1 diabetic children.34,88 Additionally, the presence of hypertension can aggravate diastolic dysfunction, as demonstrated through further and severe impairment of LV relaxation and abnormal LV filling.48 Speckle tracking echocardiography has emerged as a novel beneficial diagnostic method for early detection of LV dysfunction in diabetes, therefore being useful to detect LV abnormalities.89 This method has been shown to overcome some of the limitations of transthoracic Doppler imaging90 and can be an equally, if not better, powerful approach to assessing myocardial velocities and strain.90–92 Studies have shown it’s usefulness through examining LV strain in hypertensive and type 2 diabetic patients,93 and LV rotational mechanics in hypertensive type 2 mellitus diabetic patients.89 This echocardiography method has also been utilized in animal models of diabetes, namely assessing systolic strain and contractile function in db/db mice by Li et al.94 and assessing cardiac dysfunction in rat models of type 1 and type 2 diabetes mellitus by Matyas et al.95
Experimental evidence in animals complements these observations in human studies, showing a decrease in end-diastolic volume in alloxan diabetic dogs,96 a reduced E and A transmitral flow in db/db mice97; and an increased isovolumetric relaxation time and increased LV end-diastolic pressure in streptozotocin-induced non-insulin-dependent rats compared to controls.98 Additionally, Otsuka Long-Evans Tokushima fatty rats show increased deceleration time.99
Systolic dysfunction is also present in diabetic cardiomyopathy, although in both human and animal studies it has been shown to take longer to develop and usually occurs after diastolic dysfunction.34,97 In human studies, this manifests mainly as a reduction in ejection fraction,100,101 along with increased LV end-systolic volume100 and reduced fractional shortening.102 The Strong Heart Study showed systolic dysfunction to occur, as evidenced by lower LV fractional shortening and decreased stress corrected midwall shortening in diabetic patients.103 Animal studies are consistent with this, demonstrating impaired systolic function through lower peak-developed pressures,98 +dP/dt, peak emptying rates,104 peak filling rates, fractional shortening,105 and systolic blood pressure in streptozotocin-induced diabetic rats.
Impaired diastolic function is either associated with normal systolic function48,81,82,106,107 or can precede systolic dysfunction.34 In support of this, Raev34 showed diastolic damage and abnormalities to be more prevalent than systolic damage and abnormalities in type 1 diabetic patients, while systolic dysfunction occurred much later in the progression of diabetes. However, Fang et al. believe the use of less sensitive techniques to measure systolic dysfunction accounts for the reason behind studies demonstrating diastolic dysfunction with normal systolic function.
2.2 Cardiac hypertrophy
Diabetic cardiomyopathy is often associated with LV hypertrophy.76 The Strong Heart Study showed an independent association between diabetes and cardiac hypertrophy.103 Increased myocardial wall thickness can be seen in type 1 and 2 diabetes, alongside ventricular dysfunction.82,103,108 Additionally, myocardial hypertrophy is linked to adverse cardiovascular outcomes, including being a predictor of cardiovascular death.109,110 The Framingham study demonstrated that increased LV mass is associated with increased risk of cardiovascular outcomes of mobility and mortality.110 Moreover, Solomon et al.111 observed a slightly greater wall thickness in diabetic patients alongside decreased ventricle size, which they believe may be associated with higher filling pressures and diastolic dysfunction. These findings have been confirmed in animal studies showing an increase in LV mass alongside impaired LV relaxation and increased chamber stiffness.98 The observed LV hypertrophy in diabetics may precede the onset of systolic dysfunction, and can additionally be used as a diagnostic indicator in the development of heart failure in diabetics.112
2.3 Cardiac fibrosis
Myocardial fibrosis and collagen accumulation can manifest as a major structural alteration in the setting of diabetes,113,114 which can lead to myocardial damage and heart failure.115–118 Multiple human studies demonstrate the presence of fibrosis in the left ventricle, alongside collagen accumulation in the interstitial and perivascular region of diabetic patients.76,113,119 This cardiac fibrosis is associated with cardiac dysfunction,53,117 and may lead to worsened cardiac outcomes including developing congestive heart failure.53,116,120
Increased cardiac fibrosis in diabetes is supported by animal studies, where multiple mechanisms may be responsible for the cardiac fibrosis observed in diabetes. Otsuka Long-Evans Tokushima Fatty rats, a model of type 2 diabetes, show increased myocardial collagen content that is associated with impaired diastolic function through prolonged deceleration times and decreased early filling wave peak velocities.99 Streptozotocin-induced diabetic rats have increased collagen and interstitial fibrosis due to oxidative stress, alongside decreased cardiac contractility.121,122 Additionally, streptozotocin-induced diabetic mice show a time-dependent increase in LV collagen content, alongside impaired diastolic and systolic function.115 This is suggested to be due to reduced matrix metalloproteinase 2 levels. Spiro et al.123 also showed an increase in type IV collagen in the myocardium of diabetic rats. The increase in cardiac collagen in diabetes may be due to increased transforming growth factor-B1 (TGF-B1) receptor II expression.69,99,124 Another factor that can mediate this collagen accumulation and fibrosis development in diabetes is the myocardial accumulation of advanced glycosylation end products (AGEs)125,126 which will be discussed in more detail in a subsequent section of this review.
2.4 Impaired coronary microvascular perfusion
Abnormalities in coronary artery function and circulation are highly prevalent in diabetes,127 which may predispose the diabetic myocardium to cardiac damage and disease, including ischaemia due to impaired blood circulation and flow.127,128 Coronary flow reserve (CFR) is reduced in both type 1 and 2 diabetic patients,77,129 along with a reduction in coronary vasodilation130 due to reduced nitric oxide (NO) production.131,132 Marciano et al.133 showed that type 2 diabetic patients without coronary artery disease have impaired coronary microvascular function, demonstrated by a lower CPT-CF ratio (cold pressure test to coronary flow), compared to non-diabetic individuals. Additionally, Bagi et al.134 showed enhanced superoxide production and decreased NO production, leading to reduced coronary dilation in coronary arterioles isolated from db/db mice. Hyperglycaemia may be a cause for this, as shown by an association between CFR and HBA1c levels.135 Additionally, Durante et al.136 showed lower coronary flows in diabetic BB rats in response to stimulation by noradrenaline, calcium, or tachycardia. Coronary microvascular perfusion may be further impaired by the presence of hypertension-induced vascular lesions.137 A reduction in coronary capillary density in the diabetic myocardium has also been observed, due to lower angiogenesis as a result of decreased vascular endothelial growth factor (VEGF) expression.138–140 VEGF and VEGF receptor mRNA and protein expression were shown to be significantly decreased in diabetic and insulin-resistant non-diabetic rats,138,139 and was accompanied by decreased myocardial perfusion and LV dysfunction.139 Together, this suggests that structural abnormalities occur alongside functional abnormalities in the coronary microvasculature in diabetes. However, it is not clear which precedes the other, as some studies suggest structural changes in coronary arterial vasculature may be involved in causing further cardiac dysfunction141 and also concurrently progress as diabetic cardiomyopathy progresses.139,142 Additionally, Giordano et al.143 showed that VEGF is a critical determinant of cardiac function as a VEGF knockout mouse model resulted in contractile dysfunction. Therefore, further studies need to be done to fully elucidate the interplay in sequence of events between abnormalities in coronary capillary density and cardiac function.
Additionally, impaired CFR and vasodilation may be an early marker of atherosclerosis, which can lead to progressive deterioration of the myocardium128 and an increase in the risk of cardiovascular disease129 and ischaemia.130 Impaired vasodilation in diabetes,132 which may also be due to NO production inhibition due to hyperglycaemia144 through the generation of oxygen-derived free radicals,145 can also lead to arterial atherosclerosis. Therefore, this abnormal coronary flow may greatly increase the risk and likelihood of myocardial ischaemia.130,136,146
3. Underlying mechanisms contributing to the development of diabetic cardiomyopathy
3.1 Insulin resistance
Insulin resistance is one of the early contributing factors to the development of diabetic cardiomyopathy.147 The decreased efficacy of insulin to lower blood glucose levels occurs as a result of hyperinsulinaemia-mediated excessive insulin receptor signalling or down-regulation of insulin receptor signalling. This insulin resistance contributes to a number of adverse changes in the heart that include alterations in cardiac energy metabolism, increased inflammation and hypertrophy, lipotoxicity, glucotoxicity, alterations in mitochondrial function and reactive oxygen species (ROS) production, accumulation of advanced glycation products and O-GlcNAcylation, alterations in cardiac cardiomyocyte Ca2+ handling, systemic hyperglycaemia and hyperlipidaemia,148 and neurohormonal changes (all of which are discussed below).
It is important to note that cardiac insulin resistance precedes the development of cardiac dysfunction and heart failure. A study in mice with heart failure developed diastolic dysfunction at 2 weeks and systolic dysfunction at 3 weeks.41 Notably, the decline in function was preceded by significant cardiac insulin resistance which was determined via serial and direct measurements of insulin-stimulated glucose metabolism in isolated working hearts.41 These findings are supported by epidemiological studies that also found that insulin resistance is a predictor, rather than a biomarker, of heart failure. In an epidemioloical study of 1187 elderly men that did not have congestive heart failure, between 1990 and 1995 found that insulin resistance significantly increased the risk and predicted congestive heart failure.149 Another study in 431, 50-year-old men with a 20-year follow-up, patients that developed heart failure at age 70 presented increased plasma proinsulin at age 50, signifying that insulin resistance preceded cardiac dysfunction.150
Insulin signalling begins with insulin binding to the insulin receptor, resulting in activation of the insulin receptor substrate (IRS)-1/2, PI3K/PKB (Akt) activation, GLUT4 translocation to the cell membrane, stimulation of mitochondrial glucose oxidation, and inhibition of fatty acid oxidation.151 Cardiac muscle biopsies from type 2 diabetic patients have depressed PI3K/PKB signalling and decreased GLUT4 expression.152 In addition to decreased translocation of GLUT4, impaired PI3K engagement and stimulation of Akt also occur, due to increased phosphorylation of the serine residue on IRS-1/2.153
Activation of forkhead box-containing proteins regulates insulin signalling, leading to insulin resistance. FoxO proteins are elevated in mice with high-fat diet-induced diabetes, which down-regulates IRS1, consequently leading to decreased Akt signalling, insulin resistance and the development of diabetic cardiomyopathy.154 Also important in the regulation of insulin signalling, and is perturbed in diabetic cardiomyopathy, is the E3 ubiquitin ligase—mitsugumin 53.155 In support of this, cardiac-specific overexpression of mitsugumin 53 in mice results in severe diabetic cardiomyopathy and insulin resistance, due to degradation of the insulin receptor and IRS-1. Mitsugumin 53 overexpression is involved in transcriptionally up-regulating peroxisome proliferator-activated receptor-α (PPARα), contributing to lipid accumulation.156 This accumulation of lipid intermediates [diacylglycerol (DAG) and ceramides] contributes to the development of insulin resistance.157,158 Conversely, decreasing myocardial levels of ceramide and DAG is accompanied by improvements in insulin sensitivity and myocardial glucose utilization.159
3.2 Altered cardiac energy metabolism
The heart has a very high energy demand despite having very low ATP stores (ATP levels effectively turnover in the heart every 5–10 s).160 The heart has the ability to continually generate large amounts of ATP from various energy substrates, including fatty acids, glucose, lactate, ketone bodies and amino acids, regardless of workload, nutritional status, and hormonal status.160,161 However, this metabolic flexibility is impaired in many forms of heart disease, including diabetic cardiomyopathy.160 Insulin resistance results in an increase in myocardial fatty acid oxidation rates in diabetic cardiomyopathy and impaired glucose oxidation rates (Figure 1).162–164 This increases myocardial oxygen consumption, decreases cardiac efficiency and strongly correlates with impaired cardiac contraction and diastolic function.39,40,165–167

Figure 1
Energy metabolic changes in a healthy setting vs. diabetic cardiomyopathy. In the healthy heart, approximately 60% of the heart’s energy comes from the oxidation of fatty acids, followed by approximately 5% by glycolysis and 25% from glucose oxidation, and 10% by ketone oxidation. However, in diabetic cardiomyopathy, due to systemic and local changes in energy substrate concentrations as well as insulin resistance, the metabolic protein machinery is perturbed and subsequently, the heart’s overall energy metabolic profile is impaired. As such, diabetic cardiomyopathy results in an increase in fatty acid oxidation, decreased glucose metabolism, and decreased ketone oxidation. AGEs, advanced glycation end products; HF, heart failure; HFpEF, heart failure with preserved ejection fraction; HFrEF heart failure with reduced ejection fraction.
Multiple mechanisms contribute to the increased reliance of the heart on fatty acid use during diabetes. The first such mechanism is increased supply of fatty acids to the heart. The lack of insulin suppressive action on adipose tissue results in the release of fatty acids from adipocyte to the circulation, leading to elevation of blood plasma free fatty acid levels. Fatty acid delivery and uptake to the heart is also increased in diabetes, due to an increase in cardiac myocyte lipoprotein lipase (LPL) activity and increases in sarcolemmal CD36 protein expression, respectively.168,169 The increased uptake of fatty acids across the sarcolemma is facilitated by at least three proteins, namely, CD36, fatty acid transport protein (FATP), and fatty acid binding protein plasma membrane (FABPpm).170 In STZ-induced type 1 diabetic rats and type 2 db/db mice an up-regulation of CD36 and FABPpm protein expression occurs.171,172 While CD36 alone accounts for more than half of the total fatty acid taken up by cardiomyocytes,173 both its expression and membrane localizations are increased in diabetes.171,173
Activation of transcription regulators such as PPARs can promote expression of genes that facilities fatty acid uptake, storage and oxidation in the heart.151,174,175 Myocardial PPARα expression is increased in type 2 diabetes, and mice lacking PPARα are protected from the development of diabetic cardiomyopathy.39,176 In contrast, a recent study found no differences in the risk of cardiac dysfunction between wildtype and PPARα deficient mice subjected to a low dose of streptozotocin (STZ).177 The discrepancy in the findings could be due to the different methodology followed for inducing Type 1 diabetes (one single injection of high STZ dose vs. five daily injections of lower dose) and/or the different time points of cardiac function assessment (6 weeks vs. 9–12 weeks post-STZ administration). Interestingly, it has been shown that PPARα/γ alterations may contribute to cardiac dysfunction independent of changes in fatty acid oxidation or lipid storage in non-diabetic animals.178,179
One effect of increased PPARα in diabetes is an increase in mitochondrial carnitine palmitoyltransferase I (CPT-1) expression, a key enzyme involved in mitochondrial uptake and oxidation of fatty acids.180 In addition, perturbations in CPT-1 regulation also occur in diabetes. CPT-1 is inhibited by malonyl CoA produced from acetyl CoA by acetyl CoA carboxylase (ACC).181 Activation of AMP-activated protein kinase (AMPK) in diabetes inhibits ACC activity. Decreased ACC activity with parallel increases in malonyl-CoA decarboxylase activity182 decreases malonyl CoA levels, resulting in decreased inhibition of CPT1 and accelerated fatty acid oxidation rates (Figure 1).182–184 Post-translational modification of fatty acid oxidative enzymes also occurs in diabetes, resulting in an increase in fatty acid oxidation.185 Increased acetylation of major fatty acid metabolic enzymes due to decreased SIRT3 also leads to up-regulation of fatty acid oxidation and impaired glucose metabolism in the heart.186–188 In addition to increases in myocardial fatty acid oxidation, an increase in myocardial triacylglycerol content is seen in diabetics.189 Increased myocardial uptake of fatty acids leads to the increased accumulation of lipids and their intermediate metabolites, such as long and short fatty acyl-CoAs, and diacylglycerol (DAG).174,190 However, studies on the turnover of endogenous fatty acid in hearts from diabetic animals have generated variable results. We have shown an increased myocardial lipolysis rate in diabetic hearts irrespective of exogenous fatty acid concentration while endogenous synthesis rate remains unaffected.189 This is further supported by a 13C-NMR isotopic enrichment study in diabetic rat hearts.191 In contrast, others reported reduced or unchanged lipolysis and increased synthesis in the hearts of diabetics in the presence of high levels of exogenous free fatty acids.192,193 On the other hand, decreased levels of myocardial phospholipids is seen in diabetes together with impaired synthesis.194 While the precise contribution of altered phospholipid metabolism is less clear in diabetes, various studies have suggested the etiologic role of phospholipid (membrane lipid) metabolic dysregulation in lipotoxic cardiomyopathy and other forms of myocardial dysfunction.195,196
In contrast to the increased myocardial uptake and oxidation of fatty acids seen in diabetes, myocardial glucose transport, glycolysis and glucose oxidation are decreased in diabetes.197–200 Total myocardial GLUT4 and GLUT1 expression are decreased in diabetes.201 Decreased myocardial glycogen content along with a reduced myocardial glycogen synthesis rate and impaired glycogen synthase enzyme activity is also reported in hearts of diabetics.202–204 Insulin deficiency or resistance compromises the glucose transport and utilization in the heart. Also, the presence of excess fatty acid derivatives, such as fatty acyl CoA, DAG, and ceramide, leads to the inhibition of insulin signalling in the heart.205,206 In addition to decreased glucose transport, inhibition of cardiac phosphofructokinase (PFK-1), the rate limiting enzyme in glycolysis, is seen in diabetes.207,208 PFK-1 is inhibited allosterically by high levels of citrate, high ATP levels, and increases in NADH that are derived from increased fatty acid oxidation. Glucose oxidation is also decreased, due in part to increases in fatty acid oxidation, which inhibits the rate limiting enzyme of glucose oxidation—pyruvate dehydrogenase (PDH).164,197,209 PPARα activation also suppresses glucose uptake and utilization by increasing the expression of pyruvate dehydrogenase kinase 4 (PDK-4), which inhibits PDH.45,210
Similar trends of fatty acid and glucose metabolic shifts have been also observed in type 1 and 2 diabetic patients.57,211–214 In 31P and 1H magnetic resonance spectroscopy studies, a significant reduction in myocardial glucose utilization accompanied by reduced myocardial energetics [phosphocreatine to ATP ratio (PCr/ATP)] and increased myocardial fatty acid metabolism and triacylglycerol content is seen in type 2 diabetic patients.212,213 The increased rates of myocardial fatty acid oxidation persist even after insulin treatment in human type 2 diabetic patients.211 The levels of circulating free fatty acids are also negatively correlated with altered PCr/ATP ratios in patients with diabetes.215 Earlier studies recognized myocardial PCr/ATP ratios as a predictor of cardiovascular mortality in patients with dilated cardiomyopathy.216 Likewise, increased myocardial fatty acid utilization with a concomitant decrease in glucose utilization is seen in type 1 diabetic patients.57,214
Since fatty acids and glucose are the two important fuels for the heart, their balanced use is critical for maintaining normal contractile function. As a result, the decreased ‘metabolic flexibility’ and increased reliance of the heart on fatty acid as source of energy is associated with impaired myocardial function in diabetes.198,217–220 Enhanced fatty acid oxidation increases myocardial O2 consumption and decreases cardiac efficiency.221 Enhanced fatty acid oxidation also alters the mitochondrial NADH to NAD+ ratio and acetyl CoA levels which can further modify several intracellular signalling processes.186 Although evidence suggests a detrimental effect of increased fatty acid oxidation on heart function in diabetes, there are still opposing views on the role of glucose and fatty acid alterations and pathologic significance in non-diabetic heart failure. Relatively few studies have combined diabetes and heart failure to study the impact of energy metabolism in the hearts during diabetes. By combining high-fat feeding with pressure overload hypertrophy in mice we observed a marked decrease in insulin-stimulated glucose oxidation that was associated with both diastolic and systolic dysfunction.222,223 Furthermore, nutritional strategies that increase insulin-stimulated glucose oxidation are accompanied by a decreased severity of heart failure.222,223
The contribution of alterations in the use of other fuels such as ketone bodies and branched chain amino acids (BCAAs) is increasingly being recognized in heart failure pathogenesis.224–226 While ketone oxidation is increased in HFrEF and may be an adaptive process to maintain energy production,226,227 in diabetes myocardial ketone oxidation is impaired, and may result in a decrease in metabolic flexibility and a decrease in energy production in the heart.228,229 A decrease in BCAA oxidation in insulin resistant hearts also contributes to an impaired insulin signalling and a decrease in insulin-stimulated glucose oxidation.230–232
3.3 Cardiac lipotoxicity and glucotoxicity
Under normal circumstances, the uptake and oxidation of fatty acids are finely regulated resulting in only little myocardial lipid storage. However, in diabetes, a persistent elevation in circulating free fatty acids supplies the heart with excess fatty acids and promote accumulation of lipids in the cardiomyocytes (cardiac lipotoxicity) (Figure 2).233,234 The causative role of excess lipid accumulation in diabetic cardiomyopathy has been demonstrated using genetic or pharmacological approaches that modify uptake or oxidation of fatty acids. For instance, mice with cardiac-specific overexpression of PPARα exhibit increased uptake and utilization fatty acid and typical features of diabetic cardiomyopathy, including ventricular hypertrophy and systolic dysfunction.45 Recently, an increased activity of LPL was shown in epicardial adipose tissue from type 2 diabetic patients.235 Interestingly, elevated activity of LPL was associated with increased epicardial adipose tissue volume, suggesting increased fatty acids uptake. Of interest, deletion of CD36 or cardiac LpL rescues mice from a lipotoxic-induced cardiomyopathy caused by PPARα overexpression.236,237

Figure 2
The different stages of diabetic cardiomyopathy. AGEs, advanced glycation end products; HF, heart failure; HFpEF, heart failure with preserved ejection fraction; HFrEF heart failure with reduced ejection fraction.
In contrast, overexpression of CD36, FATP1, or acyl CoA synthetase results in lipotoxocity.238,239 This lipotoxicity correlates with diastolic dysfunction and other pathophysiological findings related to diabetic cardiomyopathy.240,241 Despite their contributing role in inducing cardiac lipotoxicity in diabetes, PPARα agonists (fibrates) are still in use clinically to treat hypertriglyceridaemia.242 In theory, lipotoxicity could arise either due to increased uptake or decreased oxidation of fatty acids. However, studies on pharmacological inhibition of FA oxidation or genetic manipulation of fatty acid oxidation enzymes revealed that decreased fatty acid oxidation does not actually lead to lipid accumulation.243 It has been hypothesized that the reduction in oxidative function may inhibit the uptake of fatty acid by feedback mechanism.244
In addition to fatty acid overload, cardiac lipotoxicity is also dependent on the type of fatty acids or lipids accumulated.244 Ceramide is one fatty acid derivative strongly associated with cardiac lipotoxicity. Inhibition of ceramide synthesis, either by deletion of serine palmitoyltransferase or pharmacologically by myriocin, results in significant metabolic and structural changes to the heart. Decreasing cardiac ceramide levels decreases heart weight, PDK4 expression, fatty acid oxidation rates and LV diameter, while improving glucose oxidation.245 DAG is another lipid derivative associated with cardiac lipotoxicity. Increased levels of DAG in the heart is associated with biochemical changes and macrovascular remodelling, indicating its possible role in the development of diabetic complications.159,246 Increased levels of both ceramide and DAG can also activate and facilitate the translocation of protein kinase C (PKC) to the cell membrane.246,247 Activation of PKC by excess lipids impairs β-adrenergic signalling in the heart by phosphorylating its receptor.248,249 Phosphorylation of the β-adrenergic receptor leads to its desensitization, resulting in reduced myocardial contractility in response to catecholamines.250 Increased PKC activity and its translocation to the cell membrane can also attenuate insulin signalling. Previous studies have shown that PKC can phosphorylate IRS-1 at its serine residue and blocks insulin-stimulated tyrosine phosphorylation and downstream Akt signalling.251,252
On the other hand, although triacylglycerol is the most abundant lipid that accumulates in the heart, studies suggest that its accumulation is not associated with toxic effects in the heart.253,254 Overall, these data demonstrate that excess fatty acid storage and utilization in the heart are detrimental to heart function, although the mechanistic link between lipid accumulation and cardiomyopathy development are not clearly defined.
In contrast to lipotoxicity, less is known about glucotoxicity. Although myocardial glucose transporters are down-regulated in diabetes, the heart can be still exposed to excess glucose. The increased extracellular glucose concentration results in the build-up of a glucose gradient for its transporter across the sarcolemma by mass action.255 As myocardial glucose oxidation is inhibited in diabetes,164,197,256 the increased glucose flux can lead to the accumulation of glycolysis intermediates and products. This imbalance can drive the diversion of glycolytic intermediates into pathological pathways in diabetes including PKC stimulation, the hexosamine pathway, the polyol pathway, and the formation of advanced glycation end products.255,257 Increased glucose uptake in GLUT4 transgenic mice also contributes to mitochondrial dysfunction via O-GlcNAcylation of the transcription factor ST1 and many electron transport chain subunits.258
3.4 Impaired mitochondrial function
The impact of heart failure and diabetes on mitochondrial bioenergetics has long been established. Perturbations in mitochondrial oxidative metabolism and mitochondrial ROS generation occur in both heart failure and diabetes.259–262 The reduction in cardiac function and efficiency along with impaired cardiac mitochondrial bioenergetics in obesity and diabetes is due to, at least in part, the excessive reliance on fatty acid oxidation and increased uncoupling protein content in these hearts, that contribute to ROS production.222,263 Many studies have proposed that ROS overload is a major culprit of diabetic cardiomyopathy.264–267 Mitochondria are a major source of ROS production, and increased fatty acid oxidation can promote ROS production.222,261,263,268–270 Excessive cardiac ROS production induces inflammation and activates many crucial mediators of pathological signalling cascades,271–278 such as PKC, apoptosis signal-regulating kinase-1, p38 mitogen-activated protein kinase, NH2-terminal Jun kinases (JNK), and JAK-STAT. Activation of these signalling cascades can contribute to the complications of diabetic cardiomyopathy.279,280 Furthermore, a recent study demonstrated that enhanced activity of Krüppel-like factor-5 is linked to an increase in oxidative stress in diabetic cardiomyopathy.177 This occurs through an up-regulation in the expression of NOX4 via direct binding to the NOX4 promoter.177 The accumulation of ROS, increased ceramide production and low mitochondrial abundance contributes to impaired cardiac function in the hearts of the diabetics.177
There have been many studies that have looked at the efficacy of antioxidants in managing diabetic cardiomyopathy.281–288 Antioxidants can mitigate ROS-mediated mitochondrial uncoupling, a characteristic of diabetic cardiomyopathy, in animal studies.289,290 Similarly, antioxidants are protective against mitochondrial ROS in the failing heart.291–293 Additionally, preclinical trials specifically targeting mitochondrial ROS respiratory complexes have been positive,294–296 but further clinical trials are necessary to confirm the efficacy of mitochondrial ROS scavenger under the contexts of both general heart failure and diabetic cardiomyopathy. Additionally, the use of Nrf2 activators and NOX inhibitor have shown to be effective in animal models,297,298 and calorie restriction can lower ROS production and UCP expression in Type II diabetic Otsuka Long-Evans Tokushima Fatty (OLETF) rats.275,299 Unfortunately, human clinical trials have failed to replicate these observations from animal models.300–303
3.5 Inflammation and hypertrophy
Diabetes leads to increases in intramyocardial inflammation, characterized by increases in cell adhesion molecules [intercellular adhesion molecule-1, vascular cell adhesion molecule 1 (VCAM-1)] and increased macrophage infiltration resulting in the release of inflammatory cytokines [interleukin (IL)-1β, IL-6, IL-18, TGF-β1, tumour necrosis factor-α (TNF-α)].267 Plasma concentrations of the cytokine acute-phase mediators, TNF-α and IL-6, are increased in the circulation in settings of impaired glucose tolerance, and thus, inflammation has been shown to be predictive for type 2 diabetes.304–306 This is due to an excess level of glucose and free fatty acids stressing both pancreatic islet cells and adipocytes, resulting in the release of pro-inflammatory cytokines and chemokines into the circulation that promote inflammation in other tissues such as the heart.307 Plasma TNF-α and IL-6 levels are increased and are associated with LV diastolic dysfunction in patients with diabetes.308
Systemic and local inflammation leads to fibrosis in the myocardium as well as hypertrophy and apoptosis at the level of the cardiomyocytes.309 An up-regulation of inflammatory signalling results in macrophage infiltration, cardiomyocyte apoptosis, hypertrophy, and a profibrotic response via extracellular matrix remodelling—all of which lead to impaired cardiac contractility and diabetic cardiomyopathy.310–312 Macrophage and lymphocyte infiltration into the cardiac cell are followed by secretion of pro-inflammatory cytokines (TNF-α, IL-6, IL-1β, TGF-β, interferon-γ), which leads to adverse cardiac remodelling.
Due to systemic accumulation of advanced glycation end products, angiotensin II and lipotoxicity, an increase in toll-like receptor 4 (TLR4) and tumour necrosis factor receptor 1 occurs in diabetes, leading to secretion of pro-inflammatory cytokines and subsequent cardiomyocyte death, hypertrophy, metabolic imbalances, contractile dysfunction, oxidative stress, and mitochondrial dysfunction.312,313 More specifically, high mobility group protein B1 mediates lipopolysaccharide binding to and activation of TLR4, resulting in downstream activation of nuclear factor-KB (NF-κB) and the NLRP3 inflammasome.314 Activation of the pleiotropic transcription factor, NF-κB, results in the transcription of genes that are pro-inflammatory [monocyte chemoattractant protein-1, Cyclooxygenase-2 (COX-2), VCAM-1], pro-hypertrophic [atrial natriuretic peptide (ANP), myosins], and pro-fibrosis (TGF-β, collagens, fibronectin).315,316
Hypertrophy follows inflammation in the heart of diabetics, since cytokines can induce cardiomyocyte hypertrophy.312,317–319 The pro-inflammatory cytokine TNF-α can activate the JNK and AKT/NF-κB pathway to promote cardiomyocyte hypertrophy.320 Activation of the NF-κB pathway can also result in cardiomyocyte growth.321 IL-1β, through IGF-1 downstream release from cardiac fibroblasts, promotes cardiomyocyte hypertrophy.322 Furthermore, IL-6 also contributes to cardiomyocyte hypertrophy through activation of the CaMKII and gp130 pathways which then activates the STAT3 pathway.323 Lastly, TGF-β can activate the TAK1-MKK3/6-p38MAPK pathway and PKC-ATF2 to promote cardiomyocyte hypertrophy.324,325
Myocardial inflammation can also lead to cardiomyocyte apoptosis which subsequently contributes to cardiac remodelling. TNF-α activates both extrinsic and intrinsic apoptotic pathways, as well as NF-κB, to promote cardiomyocyte death.326,327 Additionally, through NO synthase activation or C/EBP homologous protein (CHOP), IL-1β promotes apoptosis in cardiomyocytes.328 The NLRP3 inflammasome also induces apoptosis via caspase-1 activation.329 The inflammasome produces active caspase 1, that when activated results in cleavage of pro-IL-1β and pro-IL-18 and the production of active cytokines. In rats with high-fat diet and streptozotocin-induced diabetic cardiomyopathy, silencing of the NLRP3 inflammasome decreases the levels of IL-1β, and this observation is mirrored when silencing CMKLR1, a G-protein-coupled receptor for chemerin. Concurrent silencing of both NLRP3 and CMKLR1 potentiates the decrease in mature IL-1β, as well as the levels of pyroptosis, underlining the important role the NLRP3 inflammasome and chemerin/CMKLR1 axis plays in mediating inflammation and pyroptosis in the setting of diabetic cardiomyopathy.330
Myocardial inflammation not only results in the secretion of cytokines but also pro-fibrotic factors that activate fibroblasts and promotes cardiac fibrosis.317 TGF-β, a major cardiac pro-fibrotic cytokine, activates fibroblasts which results in the production of extracellular matrix proteins, increases collagen production, and decreases extracellular matrix degradation.310 Furthermore, IL-6 can suppress mir-29 and promote cardiac fibroblast proliferation and collagen production.331 TNF-α also similarly promotes cardiac fibrosis through WISP1 activation.332
Myocardial inflammation can also impair cardiac energy metabolism as IL-6 has been shown to impair myocardial glucose metabolism via SOCS3-dependent inhibition of IRS-1.333 Furthermore, NF-κB activation by TNF-α can inhibit PGC-1α and consequently, increase glucose metabolism via down-regulation of PDK4.334,335
Inflammation can also result in endothelial and microvascular damage, resulting in myocardial ischaemia and contributing to diastolic and systolic dysfunction in diabetic cardiomyopathy.267,317,336 Furthermore, inflammation promotes ROS generation and down-regulates SERCA2 (via IL-1β and IL-6), resulting in impaired Ca2+ handling and ultimately, diastolic dysfunction.337 Myocardial inflammation can also depress cardiac contractility as TNF-α, IL-6, IL-1β and IL-2 exert negative inotropic effects on the heart.338 Interestingly, a study in Zucker diabetic fatty rats treated with a β2-adrenergic receptor agonist decreased pro-inflammatory and pro-fibrotic responses in the heart and kidneys.339 β-arrestin can bind to the β2-adrenergic receptor and promote internalization, subsequently promoting desensitization. While this may imply a negative role in the setting of diabetic cardiomyopathy, β-arrestins have previously been reported to inhibit NF-κB activity via IκBα. As such, inhibition of NF-κB in the setting of diabetes via β-arrestin and modulation of inflammatory mediators via sympathetic nervous system regulation may offer an alternative therapeutic strategy for diabetic cardiomyopathy. Further studies that investigate the interplay between β-arrestin and NF-κB in the setting of diabetic cardiomyopathy are warranted.
Hyperinsulinaemia, via increased pancreatic production of insulin, follows insulin resistance in order to compensate for impaired cellular insulin actions. An excess of insulin can contribute to cardiomyocyte hypertrophy by acutely stimulating growth via the P13K/Akt-1 pathway.
3.6 Cardiac stiffness
Impairments in insulin signalling due to insulin resistance result in decreased GLUT4 translocation to the membrane and impaired PI3K/Akt signalling which results in decreased Ca2+-ATPase activity, consequently increasing intracellular Ca2+ levels contributing to cardiac stiffness and diastolic dysfunction.153
PI3K/Akt can activate endothelial NO synthase (eNOS), which results in an increase in NO that subsequently increases coronary vasodilation.153 However, insulin resistance decreases activation of eNOS and consequently decreases NO levels.340 Decreased NO results in impaired coronary microcirculation, due to impairments in coronary vascular smooth muscle cell relaxation.341,342 Therefore, insulin resistance alongside hyperinsulinaemia can contribute to cardiac stiffness and diastolic dysfunction.
3.7 Advanced glycation end products
Diabetes-induced chronic hyperglycaemia significantly increases the formation of AGEs in the heart.343 Protein glycation occurs after prolonged exposure to high concentrations of glucose, where amino groups of proteins bond non-enzymatically to glucose.344 A correlation exists between formation of glycosylated tissue proteins in the heart and the period of hyperglycaemia.345 Compared to non-diabetics pathologies, hearts from diabetic patients also show a higher abundance of AGE formation in the myocardium.346 A high abundance of AGEs also occurs in small intramyocardial arteries of the hearts of diabetic patients.347 This suggests that diabetes can exaggerate AGE formation and increase the susceptibility of myocardial vasculature to glycation.343 Formation of collagen cross-linking is a major determinant in the development of diabetic cardiomyopathy. Of relevance, an association between AGEs formation and decreased cardiac collagen solubility, and increased collagen III gene and protein expression, is seen in diabetes.348,349 AGE-induced increases in cross-linked collagen may lead to myocardium and arterial wall stiffness and eventually atherosclerotic plaque formation.350 Moreover, AGEs are also linked with other pathological pathways in diabetic cardiomyopathy, including oxidative stress351,352 and impaired Na+/K+-ATPase activity.353 Chronic hyperglycaemia increases both formation of AGE and expression of AGE receptors (RAGE), which in turn induces oxidative stress by activating transcription factor NFk-β.354 A strong association has been observed between increased RAGE elicited by diabetes and LV contractile dysfunction typical of diabetes cardiomyopathy, which is rescued by RAGE gene knockdown or blocking.355,356
3.8 Hexosamine biosynthesis pathway O-GlcNAcylation (O-GlcNAc)
O-GlcNAc is a post-translational modification that is responsible for regulating the activity of proteins.357 This process is initiated when N‐acetylglucosamine (GlcNAc) is attached to a serine or a threonine residue of a peptide via an O-linkage (O-GlcNAc). The substrate for O-GlcNAc is uridine diphosphate-N-acetylglucosamine (UDP-GlcNAc), which is synthesized via the hexosamine biosynthetic pathway (HBP). It has been estimated that 5% of intracellular glucose contributes to the HBP,358 although this has been debated as to whether it is an accurate estimation for cardiomyocytes.359,360 Nevertheless, O-GlcNAc levels are closely related to glucose availability.357 Glucose, after entering the cell is converted to fructose-6-phosphate (F6P) by hexokinase and isomerase. F6P is further processed to UDP-GlcNAc by four enzymatic reactions. UDP-GlcNAc is the substrate for O-GlcNAc transferase (OGT), which is responsible for catalysing O-GlcNAc to targeted proteins. Similar to other post-translational modifications, O-GlcNAc is a highly dynamic and reversible process, with the removal of O-GlcNAc from targeted proteins being accomplished by O-GlcNAcases.361
O-GlcNAc has been proposed to occur in the nucleus, cytoplasm, and mitochondria,357 as opposed to other types of glycosylation which can take place in the extracellular matrix.362 Chronic activation of the HBP is often associated with diabetic cardiomyopathy,363 as evidenced by increases in both gene expression and protein levels of Glutamine-fructose-6-phosphate transaminase (GFAT) in the myocardium of diabetic patients.364,365 Genetic modulation of Protein O-GlcNAcase (OGA) to a truncated, less effective form can exacerbate O-GlcNAc, inducing a higher chance of developing diabetes.366,367 Vascular dysfunction is a common feature of diabetes, and such dysfunction can be attributed to excess O-GlcNAc of proteins, such as transcription factor Sp1 and eNOS.368,369 Both protein levels and activity of OGT are elevated in rat aortic smooth muscle cells subjected to hyperglycaemia. Additionally, excessive O-GlcNAc can lead to improper Ca2+ handling.370 One specific target of O-GlcNAc is phospholamban, a protein regulating the function of SERCA2. Impairment of the function of phospholamban prevents the normal Ca2+ pumping after excitation from SERCA2, leading to improper contraction of heart muscle. The level of O-GlcNAc on cardiac proteins is carefully regulated by changes in OGT and OGA activity.371,372 O-GlcNAc may also affect complexes I, III, IV involved in mitochondrial respiration.371,373 O-GlcNAc may also impact ketone body metabolism by down-regulating β-hydroxybutyrate dehydrogenase mRNA levels as well as succinyl-CoA: 3-oxoacid CoA transferase protein levels.229 Of interest, ketone oxidation is decreased in the myocardium of diabetic mice.256
An increasing body of evidence suggests an increase in O-GlcNAcylation levels in diabetic cardiomyopathy. Cardiac β1-adrenoceptors (β1AR) can be modified by O-GlcNAcylation, and its signalling transduction negatively correlates with its O-GlcNAcylation level in adult rat cardiomyocytes.374 While circulating levels of N-terminal proteolytic fragment of histone deacetylase 4 (HDAC4) have been shown to be elevated in patients with diabetes, O-GlcNAcylation of HDAC4 is cardioprotective in a mouse model of diabetes.375 This cardioprotection is associated with a reduction in pathological Ca2+/calmodulin-dependent protein kinase II (CaMKII) signalling.375 O-GlcNAcylation also plays a role in regulating autophagy by modifying the synaptosomal-associated protein 29 (SNAP29).376 Increased O-GlcNAcylation of SNAP29 inhibits autophagic flux and causes further deterioration of cardiac diastolic dysfunction in STZ-induced diabetic rats.376 Furthermore, O-GlcNAcylation can modulate ionic homeostasis by targeting the activity of a number of ion channels. For example, acute hyperglycaemia can enhance K+ channel recovery via CaMKIIδ-S280 O-GlcNAcylation.377 Hyperglycaemia also increases O-GlcNAcylation of Nav1.5, which lead to the abnormal expression and distribution of Nav1.5, loss of function of the sodium channel, and prolongation of the PR/QT interval in the hearts of diabetics.378 In patients with T2D, increased O-GlcNAcylation is linked to the dynamic of glucose-induced impairment of eNOS activation in endothelial cells that could contribute to vascular dysfunction in T2D.379
3.9 Cardiac cell death pathways
Three main pathways are involved in cell death, apoptosis, necrosis, and autophagy.380 A controlled rate of apoptosis and autophagy is necessary for removing unwanted cells.267 However, in diabetes, cardiac cell death occurs at an accelerated rate.217,381,382 This is due to both a hyperactivated cellular death pathway and an impaired cellular defense mechanism.383 Cardiac apoptosis is elevated in diabetes,267,281,384,385 which is important since an apoptotic rate as low as 0.023% is sufficient to induce lethal cardiomyopathy.385 There are two main pathways of apoptosis: intrinsic or extrinsic. Intrinsic pathways can be initiated by various kinds of mitochondrial insult.380,386,387 After formation of a mitochondrial permeability transition pore (mPTP), cytochrome C leaks into the cytoplasm and assemble with Apaf-1, ATP, and procaspase-9, forming apoptosome.388,389 The final product activates the effector caspase: caspase-3, which will go on to cleave target proteins.390,391 Additionally, p53 is able to sense damage of DNA strands and up-regulate the transcription of two essential proteins: Bax and Fas.392 Bax is a pro-apoptotic protein that resides on the mitochondrial membrane,393 whereas Fas contributes to the extrinsic cellular death pathway. Fas will act on death receptors located on cellular membrane.394 Soluble extracellular protein, such as TNF-α can also bind to death receptors.326 Ligand binding initiates the assembly of multiprotein complex termed the death-inducing signalling complex (DISC), which recruits procaspase-8.395 Such movement results in procaspase-8 activation.396,397 Caspase-8 cleaves Bid, leading to the formation of the active form, truncated-Bid (T-Bid),398 that is pro-apoptotic by assisting the leak of cytochrome C into the cytoplasm. There is still debate regarding the relative contribution of apoptosis vs. necrosis to cardiomyocyte cell death in dilated cardiomyopathy.399 It has been reported that necrotic cardiomyocytes are more dominant compared to apoptotic cells in dilated cardiomyopathy and severe aortic stenosis.400,401 In line with this, irreversible opening of the mPTP also induces cell necrosis by ATP depletion, although it potentially triggers apoptosis via outer membrane rupture and cytochrome c release into the cytosol. Whether cardiomyocyte cell death by ischaemia-induced mPTP opening has a significant contribution to cardiomyocyte loss and subsequent interstitial fibrosis in diabetic hearts warrants further investigation.
The cellular death pathways described above are altered in diabetic cardiomyopathy (Figure 3). Direct exposure of high levels of glucose in myoblast H9c2 cells induces significant apoptotic cell death. The observations of hyperglycaemia increases caspase 3 activation and cytochrome C release in cardiac cells are consistent with previous findings where high levels of glucose elevated the expression of Bax and its translocation from cytosol to mitochondria-enriched heavy membrane fraction in vascular endothelial cells.402 On the other hand, the use of the caspase-3-specific inhibitor, Ac-DEVD-cmk, can suppress hyperglycaemia-induced apoptosis.402,403 Additionally, up-regulation of p53 in myocytes, due to hyperglycaemia, occurs at very early stages in the development of diabetic cardiomyopathy,404 whereas attenuation of p53 transcriptional activity by Insulin-like growth factor 1 (IGF-1) prevents myocardial apoptosis in diabetic mice.405 Besides high glucose, exposure to high levels of palmitate can also increase mitochondrial cytochrome C release, caspase-3 activation, followed by apoptotic cell death.406 The formation of ROS and reactive nitrogen species (RNS) in the heart is another critical mediator of diabetes-induced myocardial cell death.407 Both ROS and RNS may be involved in many aspects of the cell death pathway, such as activation of caspase 3, the PKC pathway, release of cytochrome C and death receptor activation.403,408,409 The antioxidant, metallothionein, can ameliorate this hyperglycaemia-induced myocardial cell death.38

Figure 3
Mechanisms that contribute to diabetes-induced heart failure. While the exact pathophysiology of the diabetic-induced heart failure still not fully defined, there are a number of mechanisms that play important roles in its occurrence. This includes mitochondrial dysfunction, cardiac insulin resistance, and impaired cardiac insulin signalling pathway, perturbed fuel use, low ATP levels, inflammation, advanced glycation end products, O-GlcNAcylation, cell death, neurohormonal mechanism, contractile proteins dysfunction, oxidative stress, gene reprogramming, lipotoxicity, glucose toxicity, and perturbed Ca2+ handling.
Unlike apoptosis, the role of autophagy in diabetic heart is still controversial. With some evidence suggesting that the induction of autophagy may convey protective effects,410 other studies proposed that excessive autophagy may accelerate the process to heart failure.411 Autophagy is believed to be impaired in diabetic heart. One major regulator of autophagy is insulin and impaired insulin signalling stimulates myocardial autophagy.411,412 Given that many different animal models have not shown the blunted myocardial autophagy in diabetes,413–415 it is surprising that there has not been any approved treatment that targets autophagy specifically. However, some available medicines, such as metformin, rapamycin and resveratrol,416–418 have been found to promote autophagy indirectly, in addition to their main mechanism of action. Recently, Mst1 (macrophage stimulating 1) was found to be responsible for dictating the cardiomyocyte towards either apoptosis or autophagy in diabetes.419
Study suggested that mitochondria-dependent, calcium overload-induced necrosis might contribute to the progression of heart failure.420 Although necrosis has been suggested to be a passive and unregulated form of cell death, targeting the pathways of necrosis has potential for treating cardio-cerebrovascular injury.421 Regulated necrosis can be classified into many categories, including but not limited to pyroptosis and ferroptosis. Both forms have been proposed to correlate with diabetic cardiomyopathy development. Pyroptosis is characterized by formation of plasma membrane pores and extracellular release of inflammatory cytokines. High glucose promoted cardiomyocytes pyroptosis by increasing ROS production.422 Elevated level of pyroptosis also induces cell death via the miR-214-3p/caspase-1/TGF-β1 pathway in diabetic mice.423 Among the many protective actions exerted by metformin, inhibition of pyroptosis by suppressing the mTOR pathway via AMPK activation, may decrease pyroptosis-induced cell death in diabetic cardiomyopathy.424 Therefore, therapies targeting pyroptosis may be an effective approach. On the other hand, ferroptosis is a newly discovered form of cell death, which can be initiated by either iron overload or oxidative stress.425 Of interest, hydrogen sulfide is an endogenous gaseous signalling molecule that is capable of inhibiting ferroptosis. One recent study proposed that treatment with the ferroptosis inhibitor ferrostatin-1 can prevent hyperglycaemia-induced ferroptosis.426
3.10 Alterations in cardiac Ca2+ handling
One of the early perturbations in Ca2+ homeostasis in diabetic cardiomyopathy that precedes LV dysfunction is a slow decay of the Ca2+ transient.427–430 Possible mechanisms that contribute to the occurrence of these disarrangements in Ca2+ handling include perturbations in the activity of SERCA2a,431,432 as well as malfunctions in Ca2+ handling proteins due to post-translational modifications, namely AGEs,433 O-GlcNacylation434 and carbonylation.281,435 Impaired Ca2+ handling between the sarcoplasmic reticulum and the mitochondria and alterations in Ca2+ influx and efflux to/from the cytosol and extracellular tissue and reduced activity of phospholamban and ryanodine receptors also contribute to the Ca2+ mishandling in diabetic cardiomyopathies.428,436–438 Ca2+ reuptake via SERCA2a is impaired in hearts from diabetic rats439–441 and ob/ob mice.442,443 Interestingly, overexpression of SERCA2a improves Ca2+ handling in animal models of diabetic cardiomyopathy.47 A recent study also demonstrated that insulin resistance impairs SERCA2a activity and cardiac function via inhibiting protein kinase B/striated muscle preferentially expressed protein kinase signalling.444 It has also been shown that oxidative stress in the hearts of diabetics could contribution to the development of diabetic cardiomyopathy via impairing SERCA2a activity,445 and that enhancing SERCA2a activity is associated with improved cardiac function in the hearts of diabetics.444–446
Diabetes is also accompanied by alterations in contractile proteins that are associated with the changes in contractile function in the diabetic heart.113,447,448 The decrease in contractile function in the diabetic heart is also positively liked to the decrease in cardiac ATPase activity.438,449,450 Along with the disturbances in cardiac ATPase proteins, it has also been shown that there are disturbances in isomyosin distribution and shifts from V1 to V3 in the diabetic heart.451–453 There are also decreases in Ca2+ sensitivity along with troponin T-band shift in the diabetic heart.454,455
3.11 Neurohormonal mechanisms
The role of the renin–angiotensin–aldosterone system and endothelin-1 system in the pathophysiology of both heart failure and diabetes has long been recognized.456–458 Diabetes is accompanied by an up-regulation of the renin–angiotensin–aldosterone pathway that causes an increase in afterload, an important contributor to cardiac remodelling in diabetic cardiomyopathy. Consistent with this, a number of animal studies have shown that inhibiting activity of the renin–angiotensin–aldosterone system limits the progression of diabetic cardiomyopathy.457,459,460 As a result, angiotensin-converting enzyme (ACE) inhibitors or angiotensin receptor antagonists are recommended to treat heart failure in diabetic and non-diabetic patients.461 Moreover, diabetes is also associated with alterations in systemic autonomic function and perturbations in cardiac rhythm.462,463 Despite the detrimental effect of these irregularities in the neurohormonal system, there are no therapeutic approaches presently used to target this system in the setting of diabetic cardiomyopathy.
3.12 Changes in cardiac gene regulation
In diabetic cardiomyopathy, there is differential expression of several genes involved in inflammation, fibrosis, insulin signalling, cell death, and metabolism (Figure 4).464,465 The advancements in microarray technology facilitates extensive gene expression profiling to uncover genetic mechanisms of diabetic cardiomyopathy and its therapeutic implication. Genes that are often dysregulated in diabetic cardiomyopathy are discussed in the respective section and are summarized in Figure 4.

Figure 4
Gene expression dysregulation in diabetic cardiomyopathy. ANP, atrial natriuretic peptide; CD36, cluster of differentiation 36; COL1A1, collagen type 1 alpha1; FABP, fatty-acid-binding proteins; GLUT1, glucose transpoter 1; GLUT4, glucose transporter 4; MCP-1, monocyte chemoattractant protein-1; MG53, mitsugumin 53; MMP2, matrix metalloproteinase-2; Mst1, macrophage stimulating 1; NF-kB, nuclear factor kappa B; PDK, pyruvate dehydrogenase kinase; PPARα, peroxisome proliferator-activated receptor; SERCA2a, sarcoplasmic- endoplasmic reticulum Ca2+ ATPase 2a; SOCS3, suppressor of cytokine signalling-3; TGF-β1, transforming growth factor beta 1; TNFα, tumour necrosis factor; VCAM-1, vascular cell adhesion molecule 1.
Studies both in type 1 and type 2 diabetes have shown abnormal cytosolic Ca2+ homeostasis and decreased SERCA2a expression in cardiomyocytes along with diminished contractile function.466 This is important in the development of diabetic cardiomyopathy, as SERCA2a gene transfer or overexpression can reduce diabetes-related contractile dysfunction, hypertrophy, and can differentially modulate the expression of genes involved in insulin signalling, glucose metabolism and cardiac remodelling.467 Genes involved in inflammation and immune response are also affected by diabetic cardiomyopathy. For instance, IL6 and STAT3 genes are up-regulated in patients with diabetic cardiomyopathy. On the other hand, down-regulation of SOCS3 (Suppressor of cytokine signalling 3) is observed in diabetic cardiomyopathy patients compared to healthy controls.468
Mitofusin 1 and 2 (Mfn1 and Mfn2) are mitochondrial dynamics proteins that controls fusion of the mitochondrial outer membrane.469 In db/db diabetic mice hearts, Mfn2 is down-regulated and contributes to an imbalance in mitochondrial dynamics. On the other hand, Mfn2 overexpression relieves diabetic cardiomyopathy by promoting mitochondrial fusion.470
Activation of PPARα expression, a transcription regulator, also occurs in diabetic cardiomyopathy.45,471 Importantly, over expression of PPARα activates genes involved in cardiac fatty acid utilization, while suppressing genes in glucose metabolic pathways. This suggests that dysregulation of PPARα expression contributes to the metabolic derangements observed in diabetic cardiomyopathy.45 Increased mitochondrial biogenesis has also been implicated in diabetic cardiomyopathy, and PPARα-dependent activation of PGC-1α may be a key driver of mitochondrial biogenic response in diabetic cardiomyopathy.471 An increase in PGC-1α gene expression occurs in hearts of db/db mice.39 In addition, the up-regulation of PPARα-dependent PGC-1α is associated with increased expression of proteins of the mitochondrial electron transport chain and oxidative phosphorylation such as nuclear receptors families NRF-1 and NRF-2 and mtDNA transcription and replication (mtTFA).471
Recently, changes in the levels of non-coding RNAs have been recognized as important mediators of altered gene expression. The largest portion of the genome consists of non-coding RNAs. Although they are not directly transcribed to protein products, these RNAs regulate the transcription and post-transcriptional processing of many proteins. These regulatory RNAs consist of microRNAs (miRNA), long non-coding RNAs (lncRNA) and circular RNAs (circRNAs).472 Over 4500 lncRNA genes, and 2000 microRNA genes has been identified in human genome alone.473 Although the function of the majority of non-coding RNAs are still unknown, mounting evidence suggests that these molecules play a significant role in a number of diseases processes and many of them are dysregulated in diabetic cardiomyopathy.464,465,474,475 Thus, their differential expression and role in diabetic cardiomyopathy pathogenesis is being actively investigated, partly because they may be potential biomarkers and therapeutic tools to treat diabetic cardiomyopathies.
LncRNAs are non-coding RNAs longer than 200 nucleotides in length. In addition to regulating other RNA functions, lncRNAs play an important role in epigenetic regulation by interacting with histone modifiers or chromatin remodellers or DNA.476 These lncRNAs also forms nucleic acid-protein complexes thereby regulating the activity or localization of these proteins or serves as a precursor for other miRNAs and circRNAs.477 Differential expression of lncRNAs has been reported in diabetic cardiomyopathy and their abnormal expression has a role in promoting or inhibiting the development of diabetes. In diabetic cardiomyopathy, there are significant alterations of a large number of lncRNAs that control apoptosis,478–480 fibrosis,481 and inflammation.478,482 Detailed data on lncRNA changes and their function are summarized in Table 1.
Table 1Alterations in miRNA, LncRNA, CircRNA, and their role in diabetic cardiomyopathy
Pathological pathway
. | Alteration
. | Altered non-coding RNAs
. | References
. |
---|
lncRNAs
. | miRNAs
. | circRNAs
. |
---|
Apoptosis | Up-regulated | MALAT1, MIAT | miRNA-1, miRNA-208a, miR-195, miR-34a, miR-483-3p | circRNA_010567, circHIPK3 | 479,491–493,502,503,605,606 |
Down-regulated | HOTAIR, LncRNA H19 | miR-29 | – | 478,480,482,494 |
Fibrosis | Up-regulated | Zfas1 | – | – | 481 |
Down-regulated | Crnde | miR-133a, miR-15a/b, miR-146a | – | 488–490,607 |
Hypertrophy | Up-regulated | – | miR-451 | – | 487 |
Down-regulated | – | miR-150, miR-373, miRNA133a, miR-30c, miR-181a | – | 483–486 |
Oxidative stress | Up-regulated | – | miR-1, miR-144 | – | 495,496 |
Inflammation | Up-regulated | – | miR-200 | – | 497 |
Down-regulated | HOTAIR | miR-146a | – | 478,490 |
Pyroptosis | Up-regulated | – | miR-30d | circ_0076631 | 504,608 |
Angiogenesis | Up-regulated | – | miR-193-5p | – | 609 |
Autophagy | Down-regulated | – | miR30c | – | 610 |
Pathological pathway
. | Alteration
. | Altered non-coding RNAs
. | References
. |
---|
lncRNAs
. | miRNAs
. | circRNAs
. |
---|
Apoptosis | Up-regulated | MALAT1, MIAT | miRNA-1, miRNA-208a, miR-195, miR-34a, miR-483-3p | circRNA_010567, circHIPK3 | 479,491–493,502,503,605,606 |
Down-regulated | HOTAIR, LncRNA H19 | miR-29 | – | 478,480,482,494 |
Fibrosis | Up-regulated | Zfas1 | – | – | 481 |
Down-regulated | Crnde | miR-133a, miR-15a/b, miR-146a | – | 488–490,607 |
Hypertrophy | Up-regulated | – | miR-451 | – | 487 |
Down-regulated | – | miR-150, miR-373, miRNA133a, miR-30c, miR-181a | – | 483–486 |
Oxidative stress | Up-regulated | – | miR-1, miR-144 | – | 495,496 |
Inflammation | Up-regulated | – | miR-200 | – | 497 |
Down-regulated | HOTAIR | miR-146a | – | 478,490 |
Pyroptosis | Up-regulated | – | miR-30d | circ_0076631 | 504,608 |
Angiogenesis | Up-regulated | – | miR-193-5p | – | 609 |
Autophagy | Down-regulated | – | miR30c | – | 610 |
Table 1Alterations in miRNA, LncRNA, CircRNA, and their role in diabetic cardiomyopathy
Pathological pathway
. | Alteration
. | Altered non-coding RNAs
. | References
. |
---|
lncRNAs
. | miRNAs
. | circRNAs
. |
---|
Apoptosis | Up-regulated | MALAT1, MIAT | miRNA-1, miRNA-208a, miR-195, miR-34a, miR-483-3p | circRNA_010567, circHIPK3 | 479,491–493,502,503,605,606 |
Down-regulated | HOTAIR, LncRNA H19 | miR-29 | – | 478,480,482,494 |
Fibrosis | Up-regulated | Zfas1 | – | – | 481 |
Down-regulated | Crnde | miR-133a, miR-15a/b, miR-146a | – | 488–490,607 |
Hypertrophy | Up-regulated | – | miR-451 | – | 487 |
Down-regulated | – | miR-150, miR-373, miRNA133a, miR-30c, miR-181a | – | 483–486 |
Oxidative stress | Up-regulated | – | miR-1, miR-144 | – | 495,496 |
Inflammation | Up-regulated | – | miR-200 | – | 497 |
Down-regulated | HOTAIR | miR-146a | – | 478,490 |
Pyroptosis | Up-regulated | – | miR-30d | circ_0076631 | 504,608 |
Angiogenesis | Up-regulated | – | miR-193-5p | – | 609 |
Autophagy | Down-regulated | – | miR30c | – | 610 |
Pathological pathway
. | Alteration
. | Altered non-coding RNAs
. | References
. |
---|
lncRNAs
. | miRNAs
. | circRNAs
. |
---|
Apoptosis | Up-regulated | MALAT1, MIAT | miRNA-1, miRNA-208a, miR-195, miR-34a, miR-483-3p | circRNA_010567, circHIPK3 | 479,491–493,502,503,605,606 |
Down-regulated | HOTAIR, LncRNA H19 | miR-29 | – | 478,480,482,494 |
Fibrosis | Up-regulated | Zfas1 | – | – | 481 |
Down-regulated | Crnde | miR-133a, miR-15a/b, miR-146a | – | 488–490,607 |
Hypertrophy | Up-regulated | – | miR-451 | – | 487 |
Down-regulated | – | miR-150, miR-373, miRNA133a, miR-30c, miR-181a | – | 483–486 |
Oxidative stress | Up-regulated | – | miR-1, miR-144 | – | 495,496 |
Inflammation | Up-regulated | – | miR-200 | – | 497 |
Down-regulated | HOTAIR | miR-146a | – | 478,490 |
Pyroptosis | Up-regulated | – | miR-30d | circ_0076631 | 504,608 |
Angiogenesis | Up-regulated | – | miR-193-5p | – | 609 |
Autophagy | Down-regulated | – | miR30c | – | 610 |
Alterations in many microRNAs (miRNAs) are also linked to changes in gene expression patterns in diabetic cardiomyopathy. MiRNAs are short non-coding RNAs that regulate gene expression by binding to the 3′ untranslated region of target messenger RNAs (mRNAs).426 Upon binding, miRNAs repress gene expression by destabilizing or degrading the target mRNAs. To date, over 2650 mature miRNAs have been identified in humans that are implicated in various diseases.427 The role of various miRNAs in mediating diabetic cardiomyopathies have been studied broadly and is summarized in Table 1. Of importance, these studies have suggested the contribution of specific miRNAs to hypertrophic,483–487 fibrotic,488–490 apoptotic,491–494 inflammatory, and oxidative stress490,495–497 changes in diabetic cardiomyopathy.
Circular RNAs (circRNAs) are produced during the processing of pre‐mRNA.473 They are involved in the regulation of pre‐mRNA splicing and RNA polymerase II.498,499 Analysis of the circRNA expression profiles in diabetic cardiomyopathy has shown differential regulation of several circRNAs in tissues from db/db mice hearts.500 Up-regulation of these circRNAs has also been shown in association with myocardial fibrosis501–503 and pyroptosis.504
Epigenetics-mediated dysregulation of gene expression also contributes to the development of diabetic cardiomyopathies. Modification of histone proteins by lysine acetylation is a major epigenetic mechanism that regulates expression of many genes. For instance, increased acetylation of cardiac histone H3 leads to increased mRNA expression of multiple cardiomyopathy-related genes, together with cardiomyocyte hypertrophy, in diabetic mice.505 Increased acetylation of histone H3 and H4 in diabetes also leads to the recruitment of inflammatory genes promoters, including TNF-α and COX-2.506 Augmented histone acetylation at promoter regions of natriuretic peptide genes is also associated with increased expressions of ANP and brain natriuretic peptide in the heart of diabetics.507 Diabetes-specific alterations in DNA methylation is also associated with altered in the phenotype of the heart in diabetes.508 This suggests that diabetes-associated epigenetic modification may be an independent risk factor for diabetic cardiomyopathy.
4. The effect of antihyperglycaemic drugs on diabetic cardiomyopathy severity
Although antihyperglycaemic drugs significantly improve glycaemic control in diabetic patients, use of these therapies does not necessarily equate to a reduced risk of developing heart failure.509,510 This highlights that lowering blood glucose alone is not sufficient to prevent diabetic cardiomyopathy development.510 However, a number of antihyperglycaemic drugs can alter the course of cardiovascular complications in the diabetic (Table 2). The impacts of these therapies on glucose and fatty acid oxidation are also summarized in Figure 5.

Figure 5
Summary figure of various antihyperglycaemic drugs and their mode of action in the context of the heart. Fatty acid, glucose, and ketone body metabolism are represented in this figure with the key modes of homeostasis regulation presented. SGLT2 inhibitors inhibit SGLT2 in the proximal tubules and thus, prevent renal glucose reabsorption, promote glycosuria, decreased insulin release, increased hepatic ketogenesis, and increased circulating blood ketone levels. These increased circulating ketones can subsequently modulate cardiac ketone oxidation rates. Metformin’s mode of action is not well understood, although it does stimulate AMPK which inhibits ACC, decreases malonyl-CoA levels and increases fatty acid metabolism. DPP4 inhibitors prevent DPP4 from inactivating GLP-1, and thus increase GLP-1 levels to potentiate insulin secretion from pancreatic beta cells. GLP-1 receptor agonists similarly increase GLP-1 levels to increase insulin secretion. Sulfonylureas bind to SUR1, and consequently the K-ATP channel closes, depolarizing the pancreatic islet cell and increasing intracellular calcium levels to promote secretion of insulin. Lastly, TZDs have widespread actions in the body but here we focus on its role in promoting glucose metabolism and improving insulin sensitivity via binding to PPARγ and promoting the transcription of genes involved in glucose uptake and metabolism.
Table 2The effects of different classes of antidiabetes drugs on the cardiovascular system and the development of diabetic cardiomyopathy
Class of the therapy
. | Main effect
. | References
. |
---|
Metformin | Beneficial | 515,611,612 |
| Neutral | 613,516 |
| Detrimental | 614 |
Sulfonylureas | Beneficial | 525 |
| Neutral | – |
| Detrimental | 518,520–524,526 |
Thiazolidinediones | Beneficial | 528 |
| Neutral | – |
| Detrimental | 528–530 |
Dipeptidyl-peptidase 4 (DPP4) inhibitors | Beneficial | 560,561,563,615,566 |
Neutral | 555,557,559,567 |
| Detrimental | 554,564,565 |
Glucagon-like peptide 1 (GLP-1) receptor agonists | Beneficial | 539,542,616,617,618 |
Neutral | 540,541,543 |
Detrimental | 544 |
Sodium-glucose transport protein 2 (SGLT2) inhibitors | Beneficial | 571,573–578 |
Neutral | |
Detrimental | |
Insulin | Beneficial | – |
| Neutral | 582,583 |
| Detrimental | – |
Class of the therapy
. | Main effect
. | References
. |
---|
Metformin | Beneficial | 515,611,612 |
| Neutral | 613,516 |
| Detrimental | 614 |
Sulfonylureas | Beneficial | 525 |
| Neutral | – |
| Detrimental | 518,520–524,526 |
Thiazolidinediones | Beneficial | 528 |
| Neutral | – |
| Detrimental | 528–530 |
Dipeptidyl-peptidase 4 (DPP4) inhibitors | Beneficial | 560,561,563,615,566 |
Neutral | 555,557,559,567 |
| Detrimental | 554,564,565 |
Glucagon-like peptide 1 (GLP-1) receptor agonists | Beneficial | 539,542,616,617,618 |
Neutral | 540,541,543 |
Detrimental | 544 |
Sodium-glucose transport protein 2 (SGLT2) inhibitors | Beneficial | 571,573–578 |
Neutral | |
Detrimental | |
Insulin | Beneficial | – |
| Neutral | 582,583 |
| Detrimental | – |
Table 2The effects of different classes of antidiabetes drugs on the cardiovascular system and the development of diabetic cardiomyopathy
Class of the therapy
. | Main effect
. | References
. |
---|
Metformin | Beneficial | 515,611,612 |
| Neutral | 613,516 |
| Detrimental | 614 |
Sulfonylureas | Beneficial | 525 |
| Neutral | – |
| Detrimental | 518,520–524,526 |
Thiazolidinediones | Beneficial | 528 |
| Neutral | – |
| Detrimental | 528–530 |
Dipeptidyl-peptidase 4 (DPP4) inhibitors | Beneficial | 560,561,563,615,566 |
Neutral | 555,557,559,567 |
| Detrimental | 554,564,565 |
Glucagon-like peptide 1 (GLP-1) receptor agonists | Beneficial | 539,542,616,617,618 |
Neutral | 540,541,543 |
Detrimental | 544 |
Sodium-glucose transport protein 2 (SGLT2) inhibitors | Beneficial | 571,573–578 |
Neutral | |
Detrimental | |
Insulin | Beneficial | – |
| Neutral | 582,583 |
| Detrimental | – |
Class of the therapy
. | Main effect
. | References
. |
---|
Metformin | Beneficial | 515,611,612 |
| Neutral | 613,516 |
| Detrimental | 614 |
Sulfonylureas | Beneficial | 525 |
| Neutral | – |
| Detrimental | 518,520–524,526 |
Thiazolidinediones | Beneficial | 528 |
| Neutral | – |
| Detrimental | 528–530 |
Dipeptidyl-peptidase 4 (DPP4) inhibitors | Beneficial | 560,561,563,615,566 |
Neutral | 555,557,559,567 |
| Detrimental | 554,564,565 |
Glucagon-like peptide 1 (GLP-1) receptor agonists | Beneficial | 539,542,616,617,618 |
Neutral | 540,541,543 |
Detrimental | 544 |
Sodium-glucose transport protein 2 (SGLT2) inhibitors | Beneficial | 571,573–578 |
Neutral | |
Detrimental | |
Insulin | Beneficial | – |
| Neutral | 582,583 |
| Detrimental | – |
4.1 Metformin
Metformin is a first-line therapy in the majority of type 2 diabetic patients. In addition to its primary role in lowering blood glucose, beneficial effects of metformin have been shown on stimulating insulin action, decreasing inflammation,511 and improving myocardial energy metabolism.512,513 However, its effect on heart failure development remains uncertain. Some studies indicated that metformin is contraindicated in diabetic patients with heart failure due to lactic acidosis.514 A recent systematic review of nine RCTs studies that examined metformin on heart failure-related outcomes in patients with or without diabetes suggest some beneficial effects of metformin, but the overall evidence were not strong enough to make a solid conclusion about metformin decreasing heart failure severity.515 Other studies have suggested that metformin therapy does not decrease the risk of heart failure development.516,517
4.2 Sulfonylureas
Sulfonylureas, especially older generation ones, increase the risk of adverse events in type 2 diabetic patients and are associated with a greater prevalence of hypoglycaemia.518,519 A meta-analysis of 115 selected trials showed that sulfonylureas are associated with increased mortality, although major adverse cardiovascular events (MACE) did not appear to be affected.520 Another meta-analysis investigating the association of metformin and sulfonylureas on both all-cause and cardiovascular mortality in type 2 diabetic patients, showed that combination therapy resulted in an increase in relative risk for cardiovascular hospitalization, as well as fatal and non-fatal events.521 Similarly, a retrospective cohort analysis investigating the addition of insulin or a sulfonylurea in diabetic patients suggests that sulfonylureas increase the risk of non-fatal cardiovascular outcomes and all-cause mortality.522 This was also seen in a metformin and sulfonylurea combination therapy study of type 2 diabetic patients, in which patients newly treated with sulfonylureas possessed a higher risk for adverse cardiovascular events.523 Recently, a meta-regression analysis of 18 studies on the risk of cardiovascular events associated with sulfonylureas found that there was an increased risk of cardiovascular mortality and events with sulfonylurea treatment.524 In a network meta-analysis, 167 327 patients were studied to evaluate the risk of cardiovascular events with different sulfonylureas. Gliclazide and glimepiride were shown to have a lower risk of both cardiovascular-related mortality and all-cause mortality vs. glibenclamide.525 Therefore, differences in the risk of mortality exist within the class of sulfonylureas. This is further reinforced by a cohort study of patients with type 2 diabetes on monotherapy with sulfonylureas, where glyburide and glimepiride did not increase the risk of adverse cardiovascular events vs. glicazide, glipizide, and tolbutamide.526
4.3 Thiazolidinediones
Thiazolidinediones (TZDs) are known to cause fluid retention and as such, can increase the risk of congestive heart failure.527 In the Pioglitazone Clinical Trial In Macrovascular Events (PROACTIVE) study, patients with type 2 diabetes and a history of macrovascular disease were randomized to receive pioglitazone or placebo.528 Pioglitazone increased heart failure hospitalization, although this was associated with less cardiac ischaemic events.528 The Diabetes Reduction Assessment With Ramipril and Rosiglitazone Medication (DREAM) study, consisting of patients with impaired fasting glucose/glucose tolerance and no known cardiovascular disease, found that while rosiglitazone reduced diabetes and the development of renal disease, it increased new-onset heart failure.529 In the Rosiglitazone Evaluated for Cardiac Outcomes and Regulation of glycaemia in Diabetes (RECORD) trial, a multi-centre open-label study with type 2 diabetic patients, rosiglitazone increased the risk of heart failure or hospitalization by over two-fold.530 As such, rosiglitazone increases the risk of heart failure and alongside other TZDs, contain serious warnings regarding the increase in fluid retention and risk of congestive heart failure.527
4.4 Glitazars
A dual PPARα and γ agonist designed to concurrently treat hyperlipidaemia and hyperglycaemia, glitazars combine the beneficial effects of agonizing both peroxisome proliferator-activated receptors. However, glitazars present a paradox and while addressing diabetic concerns with hyperlipidaemia and hyperlycaemia, they have been shown to worsen congestive heart failure in diabetic patients.531,532 Specifically, muraglitazar increases MACE, congestive heart failure and death in a review of several clinical trials that included 3725 patients.533 Another glitazar, aleglitazar, while presenting effective antidiabetic effects, also increases the risk of heart failure.534 As such, concurrent agonism of PPARα and γ results in cardiac dysfunction, which may be due to inhibition of PGC1α and mitochondrial biogenesis.178
4.5 GLP-1 receptor agonists
Glucagon-like peptide-1 (GLP-1) agonists improve glycaemic control in diabetics by mimicking GLP-1 action.535–537 These include Exenatide, a partial structural analogue of GLP-1, with other GLP-1 analogues including liraglutide, lixisenatide,538 and semaglutide.539 Some of these GLP-1 analogues have efficacy in mediating heart failure risk in diabetics, as shown by results of multiple phase III/IV large scale double-blind randomized clinical trials. The Exenatide Study of Cardiovascular Event Lowering (EXSCEL) Trial showed that exenatide in type 2 patients with cardiovascular risk did not increase their overall risk and that the incidence of MACE was not worsened.540 The Evaluation of Cardiovascular Outcomes in Patients with Type 2 Diabetes after Acute Coronary Syndrome during Treatment with Lixisenatide (EXLXA) Trial showed similar results, where lixisenatide treatment showed no effect on MACE in type 2 diabetic patients who had a recent acute coronary event.541 In contrast, the Liraglutide Effect and Action in Diabetes (LEADER) trial showed a lower risk of MACE, including the rate of the first occurrence of death from cardiovascular causes, non-fatal myocardial infarction, or non-fatal stroke in type 2 diabetic patients with high cardiovascular risk.542 Additionally, the Trial to Evaluate Cardiovascular and Other Long-Term Outcomes with Semaglutide in Subjects with Type 2 Diabetes (SUSTAIN-6) showed a significantly lower rate of cardiovascular death, non-fatal myocardial infarction, or non-fatal stroke in type 2 diabetic patients with high cardiovascular risk.542 However, the results of the Functional Impact of GLP-1 for Heart Failure Treatment (FIGHT) Trial showed no improved post-hospitalization clinical stability with liraglutide in recently hospitalized patients with established heart failure and reduced ejection fraction.543 Examination of the effect of liraglutide on ventricular function in stable chronic heart failure patients with and without diabetes also showed that liraglutide did not improve LV ejection fraction or systolic function, and was associated with an increase in heart rate and more serious cardiac adverse events.544 Combined, this calls into question the benefits of liraglutide use in preventing diabetic cardiomyopathies.
4.6 DPP4 inhibitors
Incretins-based therapy has emerged as a novel treatment approach for diabetes management, with the inhibition of dipeptidyl peptidase 4 (DPP4) being used to prevent the cleavage and inactivation of GLP-1.545 DPP4 inhibitors increase insulin secretion from pancreatic B-cells, thereby improving insulin tolerance and glucose control.546–548 Current DPP4 inhibitors include vildagliptin, sitagliptin, and saxagliptin and are similar in their efficacy in lowering HBA1C levels549 and improving glucose tolerance in diabetes.550–553 Despite the efficacy of DPP4 inhibitors in improving glycaemic control, the efficacy of DPP4 inhibitors in improving heart failure outcomes in diabetics remains unclear. The Saxagliptin Assessment of Vascular Outcomes Recorded in Patients with Diabetes Mellitus (SAVOR)-Thrombosis in Myocardial Infarction (TIMI) 53 trial (SAVOR-TIMI53) showed a significant increase in the rate of hospitalization for heart failure in type 2 diabetic patients treated with saxagliptin.554 However, the Examination of Cardiovascular Outcomes with Alogliptin versus Standard of Care (EXAMINE) trial showed non-inferiority of alogliptin to placebo on major cardiovascular events in diabetic patients with recent acute coronary syndrome.555,556 Moreover, the Trial Evaluating Cardiovascular Outcomes with Sitagliptin (TECOS) showed that sitagliptin neither improved or decreased rates of cardiovascular events such as death, myocardial infarction, stroke, or hospitalization for heart failure in type 2 diabetics with pre-existing cardiovascular disease.557,558 Other studies support the results of EXAMINE and TECOS trials.559–562 although results from meta-analyses demonstrate conflicting evidence for the effect of DPP4 inhibitors on mediating cardiovascular disease.563–565 Animals studies have also shown conflicting results on the efficacy of DPP4 inhibitors on cardiovascular disease. Sitagliptin treatment decreased LV passive stiffness and improved global LV performance in an obese type 2 diabetic mice.566 However, long-term treatment of vildagliptin showed no cardioprotective effects on cardiac function, remodelling, or infarct size in Sprague-Dawley rats subjected to myocardial infarction induced by coronary ligation.567 Combined, these studies suggest minimal beneficial effects of DPP4 inhibitors in reducing the risk of heart failure in diabetics, and support that certain DPP4 inhibitors may be safe in patients. However, the cardiovascular safety and efficacy of DPP4 inhibitors needs to be further elucidated.
4.7 SGLT2 inhibitors
Sodium glucose co-transporter 2 inhibitors (SGLT2i) prevent glucose reabsorption in the proximal tubules of the kidney, therefore increasing its secretion into the urine and improving glycaemic control.568–570 Three SGLT2i approved for clinical use include empagliflozin, dapagliflozin, and canagliflozin. Recently, large-scale clinical trials have shown cardioprotective benefits independent of its antihyperglycaemic effect in both type 2 diabetic and non-diabetic patients.571–576 The results of the Empaglifozin Cardiovascular Outcome Event Trial in Type 2 Diabetes Mellitus Patients (EMPA-REG OUTCOMES) showed a lower occurrence of death from cardiovascular causes, non-fatal myocardial infarction, or non-fatal stroke, and a reduction in overall mortality and heart failure hospitalization in empagliflozin treated type 2 diabetics patients with cardiovascular risk compared to placebo.571 The Canagliflozin Cardiovascular Assessment Study (CANVAS) and Dapagliflozin effect on Cardiovascular Events-Thrombolysis in Myocardial Infarction 58 (DECLARE-TIMI58) trials supported the results of the EMPA-REG OUTCOMES study. The CANVAS trial showed a lower risk of cardiovascular events in type 2 diabetic patients with an elevated risk of cardiovascular disease,576 while the DECLARE-TIMI58 trial showed a reduction in cardiovascular death and heart failure hospitalization in type 2 diabetic patients with or at high risk of cardiovascular disease, although it did not reduce the rate of MACE.575 Interestingly, the Dapagliflozin and Prevention of Adverse-Outcome in Heart Failure (DAPA-HF) trial showed a reduction in the risk of mortality and heart failure reduction in patients with heart failure and reduced ejection fraction with or without type 2 diabetes.573 These results are supported by the Empagliflozin Outcome Trial in Patients with Chronic Heart Failure and a Reduced Ejection Fraction (EMPEROR-Reduced) trial.574 Combined, evidence from clinical trials show a safety and efficacy of SGLT2i as a therapeutic strategy to manage diabetes and associated cardiovascular disease, heart failure, and their risk.
Studies in animal models have also demonstrated cardiovascular benefits supporting the results from the major clinical trials. Empagliflozin improves cardiac contractility, by fractional area change, and improves microvascular function in ob/ob−/− mice.577 Additionally, empagliflozin treatment attenuates cardiac fibrosis and improves haemodynamics in hypertensive rat heart failure.578 However, despite these beneficial cardiac outcomes, the exact mechanism of the cardioprotective effects of SGLT2i are unknown.228 Multiple mechanisms have been proposed including diuresis/natriuresis, improved cardiac energy metabolism,256 reduction of inflammation,579 and prevention of ischaemia reperfusion injury580 to name a few key mechanisms. Further studies are needed to fully elucidate a mechanism to explain the observed cardioprotective effects of SGLT2i in the diabetic and non-diabetic failing heart.
4.8 Insulin
While insulin is the first-line therapy to treat T1D, it is only used to manage T2D patients when oral hypoglycaemic drugs and lifestyle do not establish glycaemic control. It has been suggested that heart failure prevalence and cardiovascular mortality is increased in patient with T2D who receive insulin.581 Evaluation of the impact of insulin therapy on cardiovascular disease in diabetic patients has been the focus of a number of recent clinical trials. For example, the ORIGIN trial (Outcome Reduction With Initial Glargine Intervention) investigated glargine’s impact compared to standard care in T2D patients with high cardiovascular risk. The trial data were neutral, and the rates of incident cardiovascular outcomes were similar in the insulin-glargine and standard-care.582 In addition, the DEVOTE trial (A Trial Comparing Cardiovascular Safety of Insulin Degludec Versus Insulin Glargine in Subjects With Type 2 Diabetes at High Risk of Cardiovascular Events) compared the cardiovascular safety of degludec, ultralong acting insulin, to insulin glargine in patients with T2D and high cardiovascular risk. The study showed that degludec was non-inferior to glargine concerning the incidence of major cardiovascular events.583 While enhancing circulating insulin levels can restore cardiac insulin sensitivity in the failing heart, enhancing cardiac efficiency and reducing cardiovascular mortality, prospective studies that aim to access this possibility directly are currently lacking.
5. The effect of heart failure drugs on glycaemic control
5.1 Renin–aldosterone–angiotensin inhibitors
ACE inhibitors have been shown to improve insulin resistance and glucose intolerance via increases in GLUT4 translocation (Table 3).584 Diabetic mice treated with the ACE inhibitor temocapril show decreases in plasma glucose and insulin levels, increases in skeletal muscle glucose uptake and increases in translocation of GLUT4 to the plasma membrane.584 In a single blind, cross-over design study in type 2 diabetic patients that had arterial hypertension, the ACE inhibitor captopril increased insulin sensitivity and improved glycaemic control.585 Captopril treatment of diabetic patients also improves glucose control.586 However, while ACE inhibitor therapy improves glycaemic control effects, a case-control study in diabetic patients found that ACE inhibitors are associated with an increase in hospitalization for severe hypoglycaemia.587
Table 3The effects of different classes of heart failure therapies on glycaemic control and diabetic cardiomyopathy
Class of the therapy
. | Main effect on glycaemic control
. | References
. |
---|
Renin–angiotensin system inhibitors | Beneficial | 584–586 |
Detrimental | 587 |
Lipid-lowering agents | Beneficial | 590 |
Detrimental | 589,590,619 |
ß-receptor blockers | Beneficial | 596 |
Detrimental | 594–596 |
Aldosterone antagonists | Beneficial | 620 |
Detrimental | 621 |
Class of the therapy
. | Main effect on glycaemic control
. | References
. |
---|
Renin–angiotensin system inhibitors | Beneficial | 584–586 |
Detrimental | 587 |
Lipid-lowering agents | Beneficial | 590 |
Detrimental | 589,590,619 |
ß-receptor blockers | Beneficial | 596 |
Detrimental | 594–596 |
Aldosterone antagonists | Beneficial | 620 |
Detrimental | 621 |
Table 3The effects of different classes of heart failure therapies on glycaemic control and diabetic cardiomyopathy
Class of the therapy
. | Main effect on glycaemic control
. | References
. |
---|
Renin–angiotensin system inhibitors | Beneficial | 584–586 |
Detrimental | 587 |
Lipid-lowering agents | Beneficial | 590 |
Detrimental | 589,590,619 |
ß-receptor blockers | Beneficial | 596 |
Detrimental | 594–596 |
Aldosterone antagonists | Beneficial | 620 |
Detrimental | 621 |
Class of the therapy
. | Main effect on glycaemic control
. | References
. |
---|
Renin–angiotensin system inhibitors | Beneficial | 584–586 |
Detrimental | 587 |
Lipid-lowering agents | Beneficial | 590 |
Detrimental | 589,590,619 |
ß-receptor blockers | Beneficial | 596 |
Detrimental | 594–596 |
Aldosterone antagonists | Beneficial | 620 |
Detrimental | 621 |
5.2 Lipid-lowering agents
Statins have a propensity to induce hyperglycaemia and have been shown to cause glucose intolerance in both animals and humans. For instance, diabetic rats treated with atorvastatin or simvastatin exhibit hyperglycaemia and glucose intolerance.588 In a meta-analysis of nine trials of patients treated with statins, mean HbA1C was higher by 0.12%, indicative of a modestly increased risk for diabetes with statin treatment.589 In another meta-analysis that investigated the effect of statin therapy on HbA1C levels as well as fasting plasma glucose, statins increased HbA1C.590 Specifically, pitavastatin improved glycaemic control while atorvastatin worsened glycaemic control.590 This diabetogenic effect was also recapitulated in a national health screening cohort of non-diabetic individuals taking statins, showing that greater adherence to the use of statins, specifically atorvastatin, rosuvastatin, pitavastatin, and simvastatin, results in increases in fasting glucose levels.591
5.3 ß-blockers
β-adrenergic stimulation promotes insulin and glucagon release while α-adrenergic stimulation inhibits insulin and glucagon secretion. Therefore, β-adrenergic receptor antagonism inhibits insulin release and may worsen glycaemic control especially during hypoglycaemia. The selectivity of the ß-blocker yields distinct metabolic effects and certain ß-blockers can exacerbate hypoglycaemic episodes by delaying glucose recover time.592,593 A retrospective study that monitored glucose in patients receiving either carvedilol or a selective second-generation ß-blocker (metoprolol or atenolol) found that ß-blockers, specifically metoprolol or atenolol, increase the odds of hypoglycaemia in these hospitalized patients.594 In hypertensive diabetic patients, treatment with propranolol or metoprolol results in mean blood sugar increases of 1.0–1.5 mM.592,595 A randomized, double-blind parallel-group trial in patients with diabetes and hypertension showed that metoprolol increases mean HbA1C, but insulin sensitivity is improved with carvedilol treatment.596 Third generation non-selective β-blockers (carvedilol) possess insulin-sensitizing properties and improve glycaemic control, while second generation β1-selective (metoprolol) antagonism worsens glycaemic control.597 To underline the distinct benefits between β-blockers, non-vasodilating β-blockers (metoprolol, propranolol and atenolol) have been shown to worsen glycaemic control while vasodilating β-blockers (carvedilol, labetalol, nebivolol) improve glucose profiles.
5.4 Aldosterone antagonists
Enhanced activity of the aldosterone signalling has been implicated in the development of diabetes-induced heart failure via triggering fibrosis and insulin resistance. Treatment of dilated cardiomyopathy patients with the aldosterone antagonist spironolactone resulted in a reduced collagen accumulation in the heart and improved LV function.598 Likewise, antagonizing aldosterone can improve diastolic function and limit fibrosis in patients with hypertensive cardiomyopathy599 and metabolic syndrome.600 Of interest is that eplerenone is shown to limit biomarkers of inflammation and insulin resistance in patients with HIV.601 Aldosterone antagonists have also shown promising effects by reducing apoptosis and improving diastolic function in murine models of diabetic cardiomyopathy.602–604 The impact of aldosterone antagonism on diastolic function, cardiac insulin resistance and inflammation in patient with diabetes-induced heart failure is yet to be determine.
6. Concluding remarks
The pathophysiology of diabetes can affect the heart through multiple mechanisms that cause structural, metabolic, and functional remodelling, leading to a well-acknowledged condition called diabetic cardiomyopathy. Diabetes-induced perturbations in insulin resistance, fuel preference, ROS generation, inflammation, cell death pathways, neurohormonal mechanisms, advanced glycated end-products accumulation, lipotoxicity, glucotoxicity, and post-translational modifications contribute to the development of diabetic cardiomyopathies. Targeting these pathways is a potential therapeutic approach to lessening the likelihood of developing diabetic cardiomyopathies. A number of antidiabetic therapies can also prevent diabetic cardiomyopathy and reverse cardiac dysfunction. These advancements will help achieve personalized treatment for diabetic patients by achieving glycaemic control and managing comorbidities and limiting cardiovascular disease. Better clarity of the mechanisms involved in diabetic cardiomyopathy should lead to better therapeutics approaches to treat patients with diabetes and heart failure.
Conflict of interest: none declared.
Funding
This work was supported by the Canadian Institutes of Health Research (Foundation Grant) to G.D.L. Q.G.K. is supported by Alberta Innovates Postgraduate Fellowship in Health Innovation. K.L.H. is a student supported by the CIHR Canadian Graduate Doctoral Scholarship, the Izaak Walton Killam Memorial Scholarship, and an Alberta Innovates Graduate Studentship. S.P. is supported by the Sir Frederick Banting and Dr. Charles Best Canada Graduate Scholarship-Masters from the Canadian Institutes of Health Research and Walter H Johns Graduate Fellowship from the University of Alberta. E.B.K. is supported by a Maternal and Child Health scholarship (MatCH) programme and an Alberta Diabetes Institutes studentship.
Data availability
Data are available upon request to the corresponding author (Dr. Gary D. Lopaschuk).
References
1World Health Organization. Noncommunicable Diseases Country Profiles 2018.
2018
: World Health Organization.
2GBD 2017 Causes of Death Collaborators.
Global, regional, and national age-sex-specific mortality for 282 causes of death in 195 countries and territories, 1980-2017: a systematic analysis for the Global Burden of Disease Study 2017
.
Lancet
2018
;
392
:
1736
–
1788
.
3Tancredi
M
, Rosengren
A
, Svensson
A-M
, Kosiborod
M
, Pivodic
A
, Gudbjörnsdottir
S
, Wedel
H
, Clements
M
, Dahlqvist
S
, Lind
M.
Excess mortality among persons with type 2 diabetes
.
N Engl J Med
2015
;
373
:
1720
–
1732
.
4Kiss
Z
, Rokszin
G
, Abonyi-Toth
Z
, Jermendy
G
, Kempler
P
, Aradi
D
, Wittmann
I.
Dissimilar impact of type 2 diabetes on cardiovascular outcomes according to age categories: a nationwide population study from Hungary
.
Cardiovasc Diabetol
2018
;
17
:
107
.
5Benjamin
EJ
, Muntner
P
, Alonso
A
, Bittencourt
MS
, Callaway
CW
, Carson
AP
, Chamberlain
AM
, Chang
AR
, Cheng
S
, Das
SR
, Delling
FN
, Djousse
L
, Elkind
MSV
, Ferguson
JF
, Fornage
M
, Jordan
LC
, Khan
SS
, Kissela
BM
, Knutson
KL
, Kwan
TW
, Lackland
DT
, Lewis
TT
, Lichtman
JH
, Longenecker
CT
, Loop
MS
, Lutsey
PL
, Martin
SS
, Matsushita
K
, Moran
AE
, Mussolino
ME
, O'Flaherty
M
, Pandey
A
, Perak
AM
, Rosamond
WD
, Roth
GA
, Sampson
UKA
, Satou
GM
, Schroeder
EB
, Shah
SH
, Spartano
NL
, Stokes
A
, Tirschwell
DL
, Tsao
CW
, Turakhia
MP
, VanWagner
LB
, Wilkins
JT
, Wong
SS
, Virani
SS
; American Heart Association Council on Epidemiology and Prevention Statistics Committee and Stroke Statistics Subcommittee.
Heart Disease and Stroke Statistics-2019 update: a report from the American Heart Association
.
Circulation
2019
;
139
:
e56
–
e528
.
6Dagenais
GR
, Leong
DP
, Rangarajan
S
, Lanas
F
, Lopez-Jaramillo
P
, Gupta
R
, Diaz
R
, Avezum
A
, Oliveira
GBF
, Wielgosz
A
, Parambath
SR
, Mony
P
, Alhabib
KF
, Temizhan
A
, Ismail
N
, Chifamba
J
, Yeates
K
, Khatib
R
, Rahman
O
, Zatonska
K
, Kazmi
K
, Wei
L
, Zhu
J
, Rosengren
A
, Vijayakumar
K
, Kaur
M
, Mohan
V
, Yusufali
A
, Kelishadi
R
, Teo
KK
, Joseph
P
, Yusuf
S.
Variations in common diseases, hospital admissions, and deaths in middle-aged adults in 21 countries from five continents (PURE): a prospective cohort study
.
Lancet
2020
;
395
:
785
–
794
.
7Low Wang
CC
, Hess
CN
, Hiatt
WR
, Goldfine
AB.
Clinical Update: cardiovascular disease in diabetes mellitus: atherosclerotic cardiovascular disease and heart failure in type 2 diabetes mellitus—mechanisms, management, and clinical considerations
.
Circulation
2016
;
133
:
2459
–
2502
.
8American Diabetes Association.
Screening for diabetes
.
Diabetes Care
2002
;
25
:
s21
–
s24
.
9Kannel
WB
, McGee
DL.
Diabetes and cardiovascular disease: the Framingham study
.
JAMA
1979
;
241
:
2035
–
2038
.
10Ho
KK
, Pinsky
JL
, Kannel
WB
, Levy
D.
The epidemiology of heart failure: the Framingham Study
.
J Am Coll Cardiol
1993
;
22
:
6A
–
13A
.
11Yancy
CW
, Jessup
M
, Bozkurt
B
, Butler
J
, Casey
DE
, Drazner
MH
, Fonarow
GC
, Geraci
SA
, Horwich
T
, Januzzi
JL
, Johnson
MR
, Kasper
EK
, Levy
WC
, Masoudi
FA
, McBride
PE
, McMurray
JJV
, Mitchell
JE
, Peterson
PN
, Riegel
B
, Sam
F
, Stevenson
LW
, Tang
WHW
, Tsai
EJ
, Wilkoff
BL.
2013 ACCF/AHA guideline for the management of heart failure: a report of the American College of Cardiology Foundation/American Heart Association Task Force on Practice Guidelines
.
J Am Coll Cardiol
2013
;
62
:
e147
–
e239
.
12Cho
NH
, Shaw
JE
, Karuranga
S
, Huang
Y
, da Rocha Fernandes
JD
, Ohlrogge
AW
, Malanda
B.
IDF Diabetes Atlas: global estimates of diabetes prevalence for 2017 and projections for 2045
.
Diabetes Res Clin Pract
2018
;
138
:
271
–
281
.
13Saeedi
P
, Petersohn
I
, Salpea
P
, Malanda
B
, Karuranga
S
, Unwin
N
, Colagiuri
S
, Guariguata
L
, Motala
AA
, Ogurtsova
K
, Shaw
JE
, Bright
D
, Williams
R
, Committee
I.
Global and regional diabetes prevalence estimates for 2019 and projections for 2030 and 2045: results from the International Diabetes Federation Diabetes Atlas, 9(th) Edition
. Diabetes Res Clin Pract
2019
;
157
:
107843
.
14Amos
AF
, McCarty
DJ
, Zimmet
P.
The rising global burden of diabetes and its complications: estimates and projections to the year 2010
.
Diabet Med
1997
;
14
:
S1
–
S85
.
15King
H
, Aubert
RE
, Herman
WH.
Global burden of diabetes, 1995-2025: prevalence, numerical estimates, and projections
.
Diabetes Care
1998
;
21
:
1414
–
1431
.
16American Diabetes Association.
9. Pharmacologic approaches to glycemic treatment: standards of medical care in Diabetes-2020
.
Diabetes Care
2020
;
43
:
S98
–
S110
.
17Kannel
WB
, Hjortland
M
, Castelli
WP.
Role of diabetes in congestive heart failure: the Framingham study
.
Am J Cardiol
1974
;
34
:
29
–
34
.
18Kannel
W
, McGee
D.
Diabetes and glucose tolerance as risk factors for cardiovascular disease: the Framingham study
.
Diabetes Care
1979
;
2
:
120
–
126
.
19Gottdiener
JS
, Arnold
AM
, Aurigemma
GP
, Polak
JF
, Tracy
RP
, Kitzman
DW
, Gardin
JM
, Rutledge
JE
, Boineau
RC.
Predictors of congestive heart failure in the elderly: the Cardiovascular Health Study
.
J Am Coll Cardiol
2000
;
35
:
1628
–
1637
.
20Bell
DSH.
Heart failure—the frequent, forgotten, and often fatal complication of diabetes
.
Diabetes Care
2003
;
26
:
2433
–
2441
.
21Garcia
MJ
, McNamara
PM
, Gordon
T
, Kannel
WB.
Morbidity and mortality in diabetics in the Framingham population. Sixteen year follow-up study
.
Diabetes
1974
;
23
:
105
–
111
.
22Das
SR
, Drazner
MH
, Yancy
CW
, Stevenson
LW
, Gersh
BJ
, Dries
DL.
Effects of diabetes mellitus and ischemic heart disease on the progression from asymptomatic left ventricular dysfunction to symptomatic heart failure: a retrospective analysis from the Studies of Left Ventricular Dysfunction (SOLVD) prevention trial
.
Am Heart J
2004
;
148
:
883
–
888
.
23De Groote
P
, Lamblin
N
, Mouquet
F
, Plichon
D
, McFadden
E
, Van Belle
E
, Bauters
C.
Impact of diabetes mellitus on long-term survival in patients with congestive heart failure
.
Eur Heart J
2004
;
25
:
656
–
662
.
24Collaboration
ERF.
Diabetes mellitus, fasting glucose, and risk of cause-specific death
.
N Engl J Med
2011
;
364
:
829
–
841
.
25Haffner
SM
, Lehto
S
, Ronnemaa
T
, Pyorala
K
, Laakso
M.
Mortality from coronary heart disease in subjects with type 2 diabetes and in nondiabetic subjects with and without prior myocardial infarction
.
N Engl J Med
1998
;
339
:
229
–
234
.
26Smith
JW
, Marcus
FI
, Serokman
R.
Prognosis of patients with diabetes mellitus after acute myocardial infarction
.
Am J Cardiol
1984
;
54
:
718
–
721
.
27Malmberg
K
, Ryden
L.
Myocardial infarction in patients with diabetes mellitus
.
Eur Heart J
1988
;
9
:
259
–
264
.
28Janand-Delenne
B
, Savin
B
, Habib
G
, Bory
M
, Vague
P
, Lassmann-Vague
V.
Silent myocardial ischemia in patients with diabetes: who to screen
.
Diabetes Care
1999
;
22
:
1396
–
1400
.
29Laakso
M.
Glycemic control and the risk for coronary heart disease in patients with non-insulin-dependent diabetes mellitus. The Finnish studies
.
Ann Intern Med
1996
;
124
:
127
–
130
.
30Iribarren
C
, Karter
AJ
, Go
AS
, Ferrara
A
, Liu
JY
, Sidney
S
, Selby
JV.
Glycemic control and heart failure among adult patients with diabetes
.
Circulation
2001
;
103
:
2668
–
2673
.
31Rubler
S
, Dlugash
J
, Yuceoglu
YZ
, Kumral
T
, Branwood
AW
, Grishman
A.
New type of cardiomyopathy associated with diabetic glomerulosclerosis
.
Am J Cardiol
1972
;
30
:
595
–
602
.
32Di Bonito
P
, Cuomo
S
, Moio
N
, Sibilio
G
, Sabatini
D
, Quattrin
S
, Capaldo
B.
Diastolic dysfunction in patients with non-insulin-dependent diabetes mellitus of short duration
.
Diabet Med
1996
;
13
:
321
–
324
.
33Schannwell
CM
, Schneppenheim
M
, Perings
S
, Plehn
G
, Strauer
BE.
Left ventricular diastolic dysfunction as an early manifestation of diabetic cardiomyopathy
.
Cardiology
2002
;
98
:
33
–
39
.
34Raev
DC.
Which left ventricular function is impaired earlier in the evolution of diabetic cardiomyopathy? An echocardiographic study of young type I diabetic patients
.
Diabetes Care
1994
;
17
:
633
–
639
.
35Zhang
H
, Zhang
Y
, Li
Z
, Liu
C
, Hou
R
, Zhu
S
, Ma
N
, Zhou
L
, Liu
Y.
Left ventricular radial systolic dysfunction in diabetic patients assessed by myocardial acceleration derived from velocity vector imaging
.
J Ultrasound Med
2012
;
31
:
1179
–
1186
.
36Shimizu
I
, Yoshida
Y
, Katsuno
T
, Tateno
K
, Okada
S
, Moriya
J
, Yokoyama
M
, Nojima
A
, Ito
T
, Zechner
R
, Komuro
I
, Kobayashi
Y
, Minamino
T.
p53-induced adipose tissue inflammation is critically involved in the development of insulin resistance in heart failure
.
Cell Metab
2012
;
15
:
51
–
64
.
37Wold
LE
, Ceylan-Isik
AF
, Fang
CX
, Yang
X
, Li
SY
, Sreejayan
N
, Privratsky
JR
, Ren
J.
Metallothionein alleviates cardiac dysfunction in streptozotocin-induced diabetes: role of Ca2+ cycling proteins, NADPH oxidase, poly(ADP-Ribose) polymerase and myosin heavy chain isozyme
.
Free Radic Biol Med
2006
;
40
:
1419
–
1429
.
38Cai
L
, Wang
Y
, Zhou
G
, Chen
T
, Song
Y
, Li
X
, Kang
YJ.
Attenuation by metallothionein of early cardiac cell death via suppression of mitochondrial oxidative stress results in a prevention of diabetic cardiomyopathy
.
J Am Coll Cardiol
2006
;
48
:
1688
–
1697
.
39Buchanan
J
, Mazumder
PK
, Hu
P
, Chakrabarti
G
, Roberts
MW
, Yun
UJ
, Cooksey
RC
, Litwin
SE
, Abel
ED.
Reduced cardiac efficiency and altered substrate metabolism precedes the onset of hyperglycemia and contractile dysfunction in two mouse models of insulin resistance and obesity
.
Endocrinology
2005
;
146
:
5341
–
5349
.
40Mazumder
PK
, O'Neill
BT
, Roberts
MW
, Buchanan
J
, Yun
UJ
, Cooksey
RC
, Boudina
S
, Abel
ED.
Impaired cardiac efficiency and increased fatty acid oxidation in insulin-resistant ob/ob mouse hearts
.
Diabetes
2004
;
53
:
2366
–
2374
.
41Zhang
L
, Jaswal
JS
, Ussher
JR
, Sankaralingam
S
, Wagg
C
, Zaugg
M
, Lopaschuk
GD.
Cardiac insulin-resistance and decreased mitochondrial energy production precede the development of systolic heart failure after pressure-overload hypertrophy
.
Circ Heart Fail
2013
;
6
:
1039
–
1048
.
42Chun
L
, Junlin
Z
, Aimin
W
, Niansheng
L
, Benmei
C
, Minxiang
L.
Inhibition of ceramide synthesis reverses endothelial dysfunction and atherosclerosis in streptozotocin-induced diabetic rats
.
Diabetes Res Clin Pract
2011
;
93
:
77
–
85
.
43Rask-Madsen
C
, Li
Q
, Freund
B
, Feather
D
, Abramov
R
, Wu
IH
, Chen
K
, Yamamoto-Hiraoka
J
, Goldenbogen
J
, Sotiropoulos
KB
, Clermont
A
, Geraldes
P
, Dall'Osso
C
, Wagers
AJ
, Huang
PL
, Rekhter
M
, Scalia
R
, Kahn
CR
, King
GL.
Loss of insulin signaling in vascular endothelial cells accelerates atherosclerosis in apolipoprotein E null mice
.
Cell Metab
2010
;
11
:
379
–
389
.
44Zhou
YT
, Grayburn
P
, Karim
A
, Shimabukuro
M
, Higa
M
, Baetens
D
, Orci
L
, Unger
RH.
Lipotoxic heart disease in obese rats: implications for human obesity
.
Proc Natl Acad Sci USA
2000
;
97
:
1784
–
1789
.
45Finck
BN
, Lehman
JJ
, Leone
TC
, Welch
MJ
, Bennett
MJ
, Kovacs
A
, Han
X
, Gross
RW
, Kozak
R
, Lopaschuk
GD
, Kelly
DP.
The cardiac phenotype induced by PPARalpha overexpression mimics that caused by diabetes mellitus
.
J Clin Invest
2002
;
109
:
121
–
130
.
46Zhao
X-Y
, Hu
S-J
, Li
J
, Mou
Y
, Chen
B-P
, Xia
Q.
Decreased cardiac sarcoplasmic reticulum Ca 2+-ATPase activity contributes to cardiac dysfunction in streptozotocin-induced diabetic rats
.
J Physiol Biochem
2006
;
62
:
1
–
8
.
47Trost
SU
, Belke
DD
, Bluhm
WF
, Meyer
M
, Swanson
E
, Dillmann
WH.
Overexpression of the sarcoplasmic reticulum Ca2+-ATPase improves myocardial contractility in diabetic cardiomyopathy
.
Diabetes
2002
;
51
:
1166
–
1171
.
48Liu
JE
, Palmieri
V
, Roman
MJ
, Bella
JN
, Fabsitz
R
, Howard
BV
, Welty
TK
, Lee
ET
, Devereux
RB.
The impact of diabetes on left ventricular filling pattern in normotensive and hypertensive adults: the Strong Heart Study
.
J Am Coll Cardiol
2001
;
37
:
1943
–
1949
.
49Vasan
RS
, Levy
D.
Defining diastolic heart failure: a call for standardized diagnostic criteria
.
Circulation
2000
;
101
:
2118
–
2121
.
50Nieminen
MS
, Brutsaert
D
, Dickstein
K
, Drexler
H
, Follath
F
, Harjola
VP
, Hochadel
M
, Komajda
M
, Lassus
J
, Lopez-Sendon
JL
, Ponikowski
P
, Tavazzi
L
; EuroHeart Survey Investigators; Heart Failure Association, European Society of Cardiology.
EuroHeart Failure Survey II (EHFS II): a survey on hospitalized acute heart failure patients: description of population
.
Eur Heart J
2006
;
27
:
2725
–
2736
.
51Maisch
B
, Alter
P
, Pankuweit
S.
Diabetic cardiomyopathy-fact or fiction?
Herz
2011
;
36
:
102
–
114
.
52van Heerebeek
L
, Hamdani
N
, Falcao-Pires
I
, Leite-Moreira
AF
, Begieneman
MP
, Bronzwaer
JG
, van der Velden
J
, Stienen
GJ
, Laarman
GJ
, Somsen
A
, Verheugt
FW
, Niessen
HW
, Paulus
WJ.
Low myocardial protein kinase G activity in heart failure with preserved ejection fraction
.
Circulation
2012
;
126
:
830
–
839
.
53Falcao-Pires
I
, Hamdani
N
, Borbely
A
, Gavina
C
, Schalkwijk
CG
, van der Velden
J
, van Heerebeek
L
, Stienen
GJM
, Niessen
HWM
, Leite-Moreira
AF
, Paulus
WJ.
Diabetes mellitus worsens diastolic left ventricular dysfunction in aortic stenosis through altered myocardial structure and cardiomyocyte stiffness
.
Circulation
2011
;
124
:
1151
–
1159
.
54Montaigne
D
, Marechal
X
, Coisne
A
, Debry
N
, Modine
T
, Fayad
G
, Potelle
C
, El Arid
J-M
, Mouton
S
, Sebti
Y
, Duez
H
, Preau
S
, Remy-Jouet
I
, Zerimech
F
, Koussa
M
, Richard
V
, Neviere
R
, Edme
J-L
, Lefebvre
P
, Staels
B.
Myocardial contractile dysfunction is associated with impaired mitochondrial function and dynamics in type 2 diabetic but not in obese patients
.
Circulation
2014
;
130
:
554
–
564
.
55Savji
N
, Meijers
WC
, Bartz
TM
, Bhambhani
V
, Cushman
M
, Nayor
M
, Kizer
JR
, Sarma
A
, Blaha
MJ
, Gansevoort
RT
, Gardin
JM
, Hillege
HL
, Ji
F
, Kop
WJ
, Lau
ES
, Lee
DS
, Sadreyev
R
, van Gilst
WH
, Wang
TJ
, Zanni
MV
, Vasan
RS
, Allen
NB
, Psaty
BM
, van der Harst
P
, Levy
D
, Larson
M
, Shah
SJ
, de Boer
RA
, Gottdiener
JS
, Ho
JE.
The association of obesity and cardiometabolic traits with incident HFpEF and HFrEF
.
JACC Heart Fail
2018
;
6
:
701
–
709
.
56Rijzewijk
LJ
, van der Meer
RW
, Smit
JW
, Diamant
M
, Bax
JJ
, Hammer
S
, Romijn
JA
, de Roos
A
, Lamb
HJ.
Myocardial steatosis is an independent predictor of diastolic dysfunction in type 2 diabetes mellitus
.
J Am Coll Cardiol
2008
;
52
:
1793
–
1799
.
57Herrero
P
, Peterson
LR
, McGill
JB
, Matthew
S
, Lesniak
D
, Dence
C
, Gropler
RJ.
Increased myocardial fatty acid metabolism in patients with type 1 diabetes mellitus
.
J Am Coll Cardiol
2006
;
47
:
598
–
604
.
58Witman
MA
, Fjeldstad
AS
, McDaniel
J
, Ives
SJ
, Zhao
J
, Barrett-O'Keefe
Z
, Nativi
JN
, Stehlik
J
, Wray
DW
, Richardson
RS.
Vascular function and the role of oxidative stress in heart failure, heart transplant, and beyond
.
Hypertension
2012
;
60
:
659
–
668
.
59Li
F
, Xu
M
, Fan
Y
, Wang
Y
, Song
Y
, Cui
X
, Fu
M
, Qi
B
, Han
X
, Zhou
J
, Ge
J.
Diffuse myocardial fibrosis and the prognosis of heart failure with reduced ejection fraction in Chinese patients: a cohort study
.
Int J Cardiovasc Imaging
2020
;
36
:
671
–
689
.
60Aoki
T
, Fukumoto
Y
, Sugimura
K
, Oikawa
M
, Satoh
K
, Nakano
M
, Nakayama
M
, Shimokawa
H.
Prognostic impact of myocardial interstitial fibrosis in non-ischemic heart failure—comparison between preserved and reduced ejection fraction heart failure
.
Circ J
2011
;
75
:
2605
–
2613
.
61Gottumukkala
RV
, Lv
H
, Cornivelli
L
, Wagers
AJ
, Kwong
RY
, Bronson
R
, Stewart
GC
, Schulze
PC
, Chutkow
W
, Wolpert
HA
, Lee
RT
, Lipes
MA.
Myocardial infarction triggers chronic cardiac autoimmunity in type 1 diabetes
.
Sci Transl Med
2012
;
4
:
138ra
–
13180
.
62Campbell
DJ
, Somaratne
JB
, Jenkins
AJ
, Prior
DL
, Yii
M
, Kenny
JF
, Newcomb
AE
, Schalkwijk
CG
, Black
MJ
, Kelly
DJ.
Impact of type 2 diabetes and the metabolic syndrome on myocardial structure and microvasculature of men with coronary artery disease
.
Cardiovasc Diabetol
2011
;
10
:
80
.
63Tschope
C
, Lam
CS.
Diastolic heart failure: what we still don't know. Looking for new concepts, diagnostic approaches, and the role of comorbidities
.
Herz
2012
;
37
:
875
–
879
.
64Kawaguchi
M
, Hay
I
, Fetics
B
, Kass
DA.
Combined ventricular systolic and arterial stiffening in patients with heart failure and preserved ejection fraction: implications for systolic and diastolic reserve limitations
.
Circulation
2003
;
107
:
714
–
720
.
65Elesber
AA
, Redfield
MM
, Rihal
CS
, Prasad
A
, Lavi
S
, Lennon
R
, Mathew
V
, Lerman
LO
, Lerman
A.
Coronary endothelial dysfunction and hyperlipidemia are independently associated with diastolic dysfunction in humans
.
Am Heart J
2007
;
153
:
1081
–
1087
.
66Amir
O
, Paz
H
, Rogowski
O
, Barshai
M
, Sagiv
M
, Shnizer
S
, Reznick
AZ
, Amir
RE.
Serum oxidative stress level correlates with clinical parameters in chronic systolic heart failure patients
.
Clin Cardiol
2009
;
32
:
199
–
203
.
67Chaanine
AH
, Joyce
LD
, Stulak
JM
, Maltais
S
, Joyce
DL
, Dearani
JA
, Klaus
K
, Nair
KS
, Hajjar
RJ
, Redfield
MM.
Mitochondrial morphology, dynamics, and function in human pressure overload or ischemic heart disease with preserved or reduced ejection fraction
.
Circ Heart Fail
2019
;
12
:
e005131
.
68Gottdiener
JS
, McClelland
RL
, Marshall
R
, Shemanski
L
, Furberg
CD
, Kitzman
DW
, Cushman
M
, Polak
J
, Gardin
JM
, Gersh
BJ
, Aurigemma
GP
, Manolio
TA.
Outcome of congestive heart failure in elderly persons: influence of left ventricular systolic function. The Cardiovascular Health Study
.
Ann Intern Med
2002
;
137
:
631
–
639
.
69Westermann
D
, Rutschow
S
, Jager
S
, Linderer
A
, Anker
S
, Riad
A
, Unger
T
, Schultheiss
HP
, Pauschinger
M
, Tschope
C.
Contributions of inflammation and cardiac matrix metalloproteinase activity to cardiac failure in diabetic cardiomyopathy: the role of angiotensin type 1 receptor antagonism
.
Diabetes
2007
;
56
:
641
–
646
.
70Mancia
G
, Bombelli
M
, Facchetti
R
, Madotto
F
, Corrao
G
, Trevano
FQ
, Giannattasio
C
, Grassi
G
, Sega
R.
Long-term risk of diabetes, hypertension and left ventricular hypertrophy associated with the metabolic syndrome in a general population
.
J Hypertens
2008
;
26
:
1602
–
1611
.
71Roy
C
, Slimani
A
, de Meester
C
, Amzulescu
M
, Pasquet
A
, Vancraeynest
D
, Beauloye
C
, Vanoverschelde
JL
, Gerber
BL
, Pouleur
AC.
Associations and prognostic significance of diffuse myocardial fibrosis by cardiovascular magnetic resonance in heart failure with preserved ejection fraction
.
J Cardiovasc Magn Reson
2018
;
20
:
55
.
72Kodama
S
, Fujihara
K
, Horikawa
C
, Sato
T
, Iwanaga
M
, Yamada
T
, Kato
K
, Watanabe
K
, Shimano
H
, Izumi
T
, Sone
H.
Diabetes mellitus and risk of new-onset and recurrent heart failure: a systematic review and meta-analysis
.
ESC Heart Fail
2020
;
7
:
2146
–
2174
.
73Targher
G
, Dauriz
M
, Laroche
C
, Temporelli
PL
, Hassanein
M
, Seferovic
PM
, Drozdz
J
, Ferrari
R
, Anker
S
, Coats
A
, Filippatos
G
, Crespo-Leiro
MG
, Mebazaa
A
, Piepoli
MF
, Maggioni
AP
, Tavazzi
L
; ESC-HFA HF Long-Term Registry investigators.
In-hospital and 1-year mortality associated with diabetes in patients with acute heart failure: results from the ESC-HFA Heart Failure Long-Term Registry
.
Eur J Heart Fail
2017
;
19
:
54
–
65
.
74MacDonald
MR
, Petrie
MC
, Varyani
F
, Ostergren
J
, Michelson
EL
, Young
JB
, Solomon
SD
, Granger
CB
, Swedberg
K
, Yusuf
S
, Pfeffer
MA
, McMurray
JJ
; CHARM Investigators.
Impact of diabetes on outcomes in patients with low and preserved ejection fraction heart failure: an analysis of the Candesartan in Heart failure: assessment of Reduction in Mortality and morbidity (CHARM) programme
.
Eur Heart J
2008
;
29
:
1377
–
1385
.
75Barasch
E
, Gottdiener
JS
, Aurigemma
G
, Kitzman
DW
, Han
J
, Kop
WJ
, Tracy
RP.
Association between elevated fibrosis markers and heart failure in the elderly: the cardiovascular health study
.
Circ Heart Fail
2009
;
2
:
303
–
310
.
76Nunoda
S
, Genda
A
, Sugihara
N
, Nakayama
A
, Mizuno
S
, Takeda
R.
Quantitative approach to the histopathology of the biopsied right ventricular myocardium in patients with diabetes mellitus
.
Heart Vessels
1985
;
1
:
43
–
47
.
77Di Carli
MF
, Janisse
J
, Grunberger
G
, Ager
J.
Role of chronic hyperglycemia in the pathogenesis of coronary microvascular dysfunction in diabetes
.
J Am Coll Cardiol
2003
;
41
:
1387
–
1393
.
78From
AM
, Scott
CG
, Chen
HH.
The development of heart failure in patients with diabetes mellitus and pre-clinical diastolic dysfunction a population-based study
.
J Am Coll Cardiol
2010
;
55
:
300
–
305
.
79Shapiro
LM.
Echocardiographic features of impaired ventricular function in diabetes mellitus
.
Br Heart J
1982
;
47
:
439
–
444
.
80Boyer
JK
, Thanigaraj
S
, Schechtman
KB
, Perez
JE.
Prevalence of ventricular diastolic dysfunction in asymptomatic, normotensive patients with diabetes mellitus
.
Am J Cardiol
2004
;
93
:
870
–
875
.
81Galderisi
M.
Diastolic dysfunction and diabetic cardiomyopathy: evaluation by Doppler echocardiography
.
J Am Coll Cardiol
2006
;
48
:
1548
–
1551
.
82Park
JW
, Ziegler
AG
, Janka
HU
, Doering
W
, Mehnert
H.
Left ventricular relaxation and filling pattern in diabetic heart muscle disease: an echocardiographic study
.
Klin Wochenschr
1988
;
66
:
773
–
778
.
83Bouchard
A
, Sanz
N
, Botvinick
EH
, Phillips
N
, Heilbron
D
, Byrd
BF
, Karam
JH
, Schiller
NB.
Noninvasive assessment of cardiomyopathy in normotensive diabetic-patients between 20 and 50 years old
.
Am J Med
1989
;
87
:
160
–
166
.
84Shimizu
M
, Sugihara
N
, Kita
Y
, Shimizu
K
, Shibayama
S
, Takeda
R.
Increase in left ventricular chamber stiffness in patients with non-insulin dependent diabetes mellitus
.
Jpn Circ J
1991
;
55
:
657
–
664
.
85Giampietro
O
, Di Bello
V
, Matteucci
E
, Talarico
L
, Ruberti
F
, Boldrini
E
, Giorgi
D
, Giusti
C.
Erythrocyte Na+/H+ exchange and preclinical abnormalities of the left ventricular diastolic function in normotensive type 1 (insulin-dependent) diabetic patients
.
Acta Diabetol
1997
;
34
:
223
–
229
.
86Ruddy
TD
, Shumak
SL
, Liu
PP
, Barnie
A
, Seawright
SJ
, McLaughlin
PR
, Zinman
B.
The relationship of cardiac diastolic dysfunction to concurrent hormonal and metabolic status in type I diabetes mellitus
.
J Clin Endocrinol Metab
1988
;
66
:
113
–
118
.
87Attali
JR
, Sachs
RN
, Valensi
P
, Palsky
D
, Tellier
P
, Vulpillat
M
, Lanfranchi
J
, Sebaoun
J.
Asymptomatic diabetic cardiomyopathy: a noninvasive study
.
Diabetes Res Clin Pract
1988
;
4
:
183
–
190
.
88Cerutti
F
, Vigo
A
, Sacchetti
C
, Bessone
A
, Barattia
G
, Morello
M
, Casalucci
D
, Gastaldi
L.
Evaluation of left ventricular diastolic function in insulin dependent diabetic children by M-mode and Doppler echocardiography
.
Panminerva Med
1994
;
36
:
109
–
114
.
89Gökdeniz
T
, Kalaycıoğlu
E
, Aykan
AÇ
, Gül
İ
, Boyacı
F
, Gürsoy
MO.
Effects of non-dipper blood pressure pattern on left ventricular rotational mechanics in hypertensive patients with type 2 diabetes mellitus: a speckle tracking study
.
Int J Cardiovasc Imaging
2014
;
30
:
57
–
65
.
90Blessberger
H
, Binder
T.
Non-invasive imaging: two dimensional speckle tracking echocardiography: basic principles
.
Heart
2010
;
96
:
716
–
722
.
91Bauer
M
, Cheng
S
, Jain
M
, Ngoy
S
, Theodoropoulos
C
, Trujillo
A
, Lin
FC
, Liao
R.
Echocardiographic speckle-tracking based strain imaging for rapid cardiovascular phenotyping in mice
.
Circ Res
2011
;
108
:
908
–
916
.
92Biswas
M
, Sudhakar
S
, Nanda
NC
, Buckberg
G
, Pradhan
M
, Roomi
AU
, Gorissen
W
, Houle
H.
Two- and three-dimensional speckle tracking echocardiography: clinical applications and future directions
.
Echocardiography
2013
;
30
:
88
–
105
.
93Mondillo
S
, Cameli
M
, Caputo
ML
, Lisi
M
, Palmerini
E
, Padeletti
M
, Ballo
P.
Early detection of left atrial strain abnormalities by speckle-tracking in hypertensive and diabetic patients with normal left atrial size
.
J Am Soc Echocardiogr
2011
;
24
:
898
–
908
.
94Li
RJ
, Yang
J
, Yang
Y
, Ma
N
, Jiang
B
, Sun
QW
, Li
YJ.
Speckle tracking echocardiography in the diagnosis of early left ventricular systolic dysfunction in type II diabetic mice
.
BMC Cardiovasc Disord
2014
;
14
:
141
.
95Matyas
C
, Kovacs
A
, Nemeth
BT
, Olah
A
, Braun
S
, Tokodi
M
, Barta
BA
, Benke
K
, Ruppert
M
, Lakatos
BK
, Merkely
B
, Radovits
T.
Comparison of speckle-tracking echocardiography with invasive hemodynamics for the detection of characteristic cardiac dysfunction in type-1 and type-2 diabetic rat models
.
Cardiovasc Diabetol
2018
;
17
:
13
.
96Regan
TJ
, Ettinger
PO
, Khan
MI
, Jesrani
MU
, Lyons
MM
, Oldewurtel
HA
, Weber
M.
Altered myocardial-function and metabolism in chronic diabetes-mellitus without ischemia in dogs
.
Circ Res
1974
;
35
:
222
–
237
.
97Semeniuk
LM
, Kryski
AJ
, Severson
DL.
Echocardiographic assessment of cardiac function in diabetic db/db and transgenic db/db-hGLUT4 mice
.
Am J Physiol Heart Circ Physiol
2002
;
283
:
H976
–
982
.
98Joffe
II
, Travers
KE
, Perreault-Micale
CL
, Hampton
T
, Katz
SE
, Morgan
JP
, Douglas
PS.
Abnormal cardiac function in the streptozotocin-induced non-insulin-dependent diabetic rat: noninvasive assessment with Doppler echocardiography and contribution of the nitric oxide pathway
.
J Am Coll Cardiol
1999
;
34
:
2111
–
2119
.
99Mizushige
K
, Yao
L
, Noma
T
, Kiyomoto
H
, Yu
Y
, Hosomi
N
, Ohmori
K
, Matsuo
H.
Alteration in left ventricular diastolic filling and accumulation of myocardial collagen at insulin-resistant prediabetic stage of a type II diabetic rat model
.
Circulation
2000
;
101
:
899
–
907
.
100Friedman
NE
, Levitsky
LL
, Edidin
DV
, Vitullo
DA
, Lacina
SJ
, Chiemmongkoltip
P.
Echocardiographic evidence for impaired myocardial performance in children with type I diabetes mellitus
.
Am J Med
1982
;
73
:
846
–
850
.
101Iwasaka
T
, Takahashi
N
, Nakamura
S
, Sugiura
T
, Tarumi
N
, Kimura
Y
, Okubo
N
, Taniguchi
H
, Matsui
Y
, Inada
M.
Residual left ventricular pump function after acute myocardial infarction in NIDDM patients
.
Diabetes Care
1992
;
15
:
1522
–
1526
.
102Shapiro
LM
, Howat
AP
, Calter
MM.
Left ventricular function in diabetes mellitus. I: methodology, and prevalence and spectrum of abnormalities
.
Br Heart J
1981
;
45
:
122
–
128
.
103Devereux
RB
, Roman
MJ
, Paranicas
M
, O’Grady
MJ
, Lee
ET
, Welty
TK
, Fabsitz
RR
, Robbins
D
, Rhoades
ER
, Howard
BV.
Impact of diabetes on cardiac structure and function: the strong heart study
.
Circulation
2000
;
101
:
2271
–
2276
.
104Hoit
BD
, Castro
C
, Bultron
G
, Knight
S
, Matlib
MA.
Noninvasive evaluation of cardiac dysfunction by echocardiography in streptozotocin-induced diabetic rats
.
J Card Fail
1999
;
5
:
324
–
333
.
105Hayashi
K
, Okumura
K
, Matsui
H
, Murase
K
, Kamiya
H
, Saburi
Y
, Numaguchi
Y
, Toki
Y
, Hayakawa
T.
Involvement of 1,2-diacylglycerol in improvement of heart function by etomoxir in diabetic rats
.
Life Sci
2001
;
68
:
1515
–
1526
.
106Rajan
S
, Gokhale
SM.
Cardiovascular function in patients with insulin‐dependent diabetes mellitus: a study using noninvasive methods
.
Ann N Y Acad Sci
2002
;
958
:
425
–
430
.
107Poirier
P
, Bogaty
P
, Garneau
C
, Marois
L
, Dumesnil
JG.
Diastolic dysfunction in normotensive men with well-controlled type 2 diabetes: importance of maneuvers in echocardiographic screening for preclinical diabetic cardiomyopathy
.
Diabetes Care
2001
;
24
:
5
–
10
.
108Carugo
S
, Giannattasio
C
, Calchera
I
, Paleari
F
, Gorgoglione
MG
, Grappiolo
A
, Gamba
P
, Rovaris
G
, Failla
M
, Mancia
G.
Progression of functional and structural cardiac alterations in young normotensive uncomplicated patients with type 1 diabetes mellitus
.
J Hypertens
2001
;
19
:
1675
–
1680
.
109Koren
MJ
, Devereux
RB
, Casale
PN
, Savage
DD
, Laragh
JH.
Relation of left ventricular mass and geometry to morbidity and mortality in uncomplicated essential hypertension
.
Ann Intern Med
1991
;
114
:
345
–
352
.
110Levy
D
, Garrison
RJ
, Savage
DD
, Kannel
WB
, Castelli
WP.
Prognostic implications of echocardiographically determined left ventricular mass in the Framingham Heart Study
.
N Engl J Med
1990
;
322
:
1561
–
1566
.
111Solomon
SD
, Sutton
MSJ
, Lamas
GA
, Plappert
T
, Rouleau
JL
, Skali
H
, Moyé
L
, Braunwald
E
, Pfeffer
MA.
Ventricular remodeling does not accompany the development of heart failure in diabetic patients after myocardial infarction
.
Circulation
2002
;
106
:
1251
–
1255
.
112Paulus
WJ
, Tschöpe
C
, Sanderson
JE
, Rusconi
C
, Flachskampf
FA
, Rademakers
FE
, Marino
P
, Smiseth
OA
, De Keulenaer
G
, Leite-Moreira
AF
, Borbély
A
, Édes
I
, Handoko
ML
, Heymans
S
, Pezzali
N
, Pieske
B
, Dickstein
K
, Fraser
AG
, Brutsaert
DL.
How to diagnose diastolic heart failure: a consensus statement on the diagnosis of heart failure with normal left ventricular ejection fraction by the Heart Failure and Echocardiography Associations of the European Society of Cardiology
.
Eur Heart J
2007
;
28
:
2539
–
2550
.
113Regan
TJ
, Lyons
MM
, Ahmed
SS
, Levinson
GE
, Oldewurtel
HA
, Ahmad
MR
, Haider
B.
Evidence for cardiomyopathy in familial diabetes mellitus
.
J Clin Invest
1977
;
60
:
884
–
899
.
114Kawaguchi
M
, Techigawara
M
, Ishihata
T
, Asakura
T
, Saito
F
, Maehara
K
, Maruyama
Y.
A comparison of ultrastructural changes on endomyocardial biopsy specimens obtained from patients with diabetes mellitus with and without hypertension
.
Heart Vessels
1997
;
12
:
267
–
274
.
115Linthout
S
, Seeland
U
, Riad
A
, Eckhardt
O
, Hohl
M
, Dhayat
N
, Richter
U
, Fischer
JW
, Böhm
M
, Pauschinger
M
, Schultheiss
H-P
, Tschöpe
C.
Reduced MMP-2 activity contributes to cardiac fibrosis in experimental diabetic cardiomyopathy
.
Basic Res Cardiol
2008
;
103
:
319
–
327
.
116van Hoeven
KH
, Factor
SM.
A comparison of the pathological spectrum of hypertensive, diabetic, and hypertensive-diabetic heart disease
.
Circulation
1990
;
82
:
848
–
855
.
117Polyakova
V
, Hein
S
, Kostin
S
, Ziegelhoeffer
T
, Schaper
J.
Matrix metalloproteinases and their tissue inhibitors in pressure-overloaded human myocardium during heart failure progression
.
J Am Coll Cardiol
2004
;
44
:
1609
–
1618
.
118Yue
Y
, Meng
K
, Pu
Y
, Zhang
X.
Transforming growth factor beta (TGF-beta) mediates cardiac fibrosis and induces diabetic cardiomyopathy
.
Diabetes Res Clin Pract
2017
;
133
:
124
–
130
.
119Shimizu
M
, Umeda
K
, Sugihara
N
, Yoshio
H
, Ino
H
, Takeda
R
, Okada
Y
, Nakanishi
I.
Collagen remodelling in myocardia of patients with diabetes
.
J Clin Pathol
1993
;
46
:
32
–
36
.
120Lovic
D
, Erdine
S
, Catakoğlu
AB.
How to estimate left ventricular hypertrophy in hypertensive patients
.
Anadolu Kardiyol Derg
2014
;
14
:
389
–
395
.
121Aragno
M
, Mastrocola
R
, Alloatti
G
, Vercellinatto
I
, Bardini
P
, Geuna
S
, Catalano
MG
, Danni
O
, Boccuzzi
G.
Oxidative stress triggers cardiac fibrosis in the heart of diabetic rats
.
Endocrinology
2008
;
149
:
380
–
388
.
122Riva
E
, Andreoni
G
, Bianchi
R
, Latini
R
, Luvara
G
, Jeremic
G
, Traquandi
C
, Tuccinardi
L.
Changes in diastolic function and collagen content in normotensive and hypertensive rats with long-term streptozotocin-induced diabetes
.
Pharmacol Res
1998
;
37
:
233
–
240
.
123Spiro
MJ
, Kumar
BRR
, Crowley
TJ.
Myocardial glycoproteins in diabetes—type-Vi collagen is a major pas-reactive extracellular-matrix protein
.
J Mol Cell Cardiol
1992
;
24
:
397
–
410
.
124Lee
AA
, Dillmann
WH
, McCulloch
AD
, Villarreal
FJ.
Angiotensin II stimulates the autocrine production of transforming growth factor-β1 in adult rat cardiac fibroblasts
.
J Mol Cell Cardiol
1995
;
27
:
2347
–
2357
.
125Asif
M
, Egan
J
, Vasan
S
, Jyothirmayi
GN
, Masurekar
MR
, Lopez
S
, Williams
C
, Torres
RL
, Wagle
D
, Ulrich
P
, Cerami
A
, Brines
M
, Regan
TJ.
An advanced glycation endproduct cross-link breaker can reverse age-related increases in myocardial stiffness
.
Proc Natl Acad Sci USA
2000
;
97
:
2809
–
2813
.
126Tang
M
, Zhong
M
, Shang
Y
, Lin
H
, Deng
J
, Jiang
H
, Lu
H
, Zhang
Y
, Zhang
W.
Differential regulation of collagen types I and III expression in cardiac fibroblasts by AGEs through TRB3/MAPK signaling pathway
.
Cell Mol Life Sci
2008
;
65
:
2924
–
2932
.
127Strauer
BE
, Motz
W
, Vogt
M
, Schwartzkopff
B.
Impaired coronary flow reserve in NIDDM: a possible role for diabetic cardiopathy in humans
.
Diabetes
1997
;
46
:
S119
–
124
.
128Nitenberg
A
, Valensi
P
, Sachs
R
, Dali
M
, Aptecar
E
, Attali
JR.
Impairment of coronary vascular reserve and ACh-induced coronary vasodilation in diabetic patients with angiographically normal coronary arteries and normal left ventricular systolic function
.
Diabetes
1993
;
42
:
1017
–
1025
.
129Pitkanen
O-P
, Nuutila
P
, Raitakari
OT
, Ronnemaa
T
, Koskinen
PJ
, Iida
H
, Lehtimaki
TJ
, Laine
HK
, Takala
T
, Viikari
JSA
, Knuuti
J.
Coronary flow reserve is reduced in young men with IDDM
.
Diabetes
1998
;
47
:
248
–
254
.
130Nahser
PJ
Jr, Brown
RE
, Oskarsson
H
, Winniford
MD
, Rossen
JD.
Maximal coronary flow reserve and metabolic coronary vasodilation in patients with diabetes mellitus
.
Circulation
1995
;
91
:
635
–
640
.
131Okon
EB
, Chung
AW
, Rauniyar
P
, Padilla
E
, Tejerina
T
, McManus
BM
, Luo
H
, van Breemen
C.
Compromised arterial function in human type 2 diabetic patients
.
Diabetes
2005
;
54
:
2415
–
2423
.
132Williams
SB
, Cusco
JA
, Roddy
MA
, Johnstone
MT
, Creager
MA.
Impaired nitric oxide-mediated vasodilation in patients with non-insulin-dependent diabetes mellitus
.
J Am Coll Cardiol
1996
;
27
:
567
–
574
.
133Marciano
C
, Galderisi
M
, Gargiulo
P
, Acampa
W
, D’Amore
C
, Esposito
R
, Capasso
E
, Savarese
G
, Casaretti
L
, Iudice
FL
, Esposito
G
, Rengo
G
, Leosco
D
, Cuocolo
A
, Perrone-Filardi
P.
Effects of type 2 diabetes mellitus on coronary microvascular function and myocardial perfusion in patients without obstructive coronary artery disease
.
Eur J Nucl Med Mol Imaging
2012
;
39
:
1199
–
1206
.
134Bagi
Z
, Koller
A
, Kaley
G.
Superoxide-NO interaction decreases flow- and agonist-induced dilations of coronary arterioles in Type 2 diabetes mellitus
.
Am J Physiol Heart Circ Physiol
2003
;
285
:
H1404
–
H1410
.
135Yokoyama
I
, Momomura
S
, Ohtake
T
, Yonekura
K
, Nishikawa
J
, Sasaki
Y
, Omata
M.
Reduced myocardial flow reserve in non-insulin-dependent diabetes mellitus
.
J Am Coll Cardiol
1997
;
30
:
1472
–
1477
.
136Durante
W
, Sunahara
FA
, Sen
AK.
Effect of diabetes on metabolic coronary dilatation in the rat
.
Cardiovasc Res
1989
;
23
:
40
–
45
.
137Candido
R
, Jandeleit-Dahm
KA
, Cao
Z
, Nesteroff
SP
, Burns
WC
, Twigg
SM
, Dilley
RJ
, Cooper
ME
, Allen
TJ.
Prevention of accelerated atherosclerosis by angiotensin-converting enzyme inhibition in diabetic apolipoprotein E-deficient mice
.
Circulation
2002
;
106
:
246
–
253
.
138Chou
E
, Suzuma
I
, Way
KJ
, Opland
D
, Clermont
AC
, Naruse
K
, Suzuma
K
, Bowling
NL
, Vlahos
CJ
, Aiello
LP
, King
GL.
Decreased cardiac expression of vascular endothelial growth factor and its receptors in insulin-resistant and diabetic States: a possible explanation for impaired collateral formation in cardiac tissue
.
Circulation
2002
;
105
:
373
–
379
.
139Yoon
YS
, Uchida
S
, Masuo
O
, Cejna
M
, Park
JS
, Gwon
HC
, Kirchmair
R
, Bahlman
F
, Walter
D
, Curry
C
, Hanley
A
, Isner
JM
, Losordo
DW.
Progressive attenuation of myocardial vascular endothelial growth factor expression is a seminal event in diabetic cardiomyopathy—restoration of microvascular homeostasis and recovery of cardiac function in diabetic cardiomyopathy after replenishment of local vascular endothelial growth factor
.
Circulation
2005
;
111
:
2073
–
2085
.
140Khazaei
M
, Fallahzadeh
AR
, Sharifi
MR
, Afsharmoghaddam
N
, Javanmard
SH
, Salehi
E.
Effects of diabetes on myocardial capillary density and serum angiogenesis biomarkers in male rats
.
Clinics (Sao Paulo)
2011
;
66
:
1419
–
1424
.
141Neglia
D
, Parodi
O
, Gallopin
M
, Sambuceti
G
, Giorgetti
A
, Pratali
L
, Salvadori
P
, Michelassi
C
, Lunardi
M
, Pelosi
G
, Marzilli
M
, L’Abbate
A.
Myocardial blood-flow response to pacing tachycardia and to dipyridamole infusion in patients with dilated cardiomyopathy without overt heart-failure—a quantitative assessment by Positron Emission Tomography
.
Circulation
1995
;
92
:
796
–
804
.
142Yarom
R
, Zirkin
H
, Stammler
G
, Rose
AG.
Human coronary microvessels in diabetes and ischaemia. Morphometric study of autopsy material
.
J Pathol
1992
;
166
:
265
–
270
.
143Giordano
FJ
, Gerber
HP
, Williams
SP
, VanBruggen
N
, Bunting
S
, Ruiz-Lozano
P
, Gu
YS
, Nath
AK
, Huang
Y
, Hickey
R
, Dalton
N
, Peterson
KL
, Ross
J
, Chien
KR
, Ferrara
N.
A cardiac myocyte vascular endothelial growth factor paracrine pathway is required to maintain cardiac function
.
Proc Natl Acad Sci USA
2001
;
98
:
5780
–
5785
.
144Angulo
J
, Rodríguez-Mañas
L
, Peiró
C
, Neira
M
, Marín
J
, Sánchez-Ferrer
CF.
Impairment of nitric oxide-mediated relaxations in anaesthetized autoperfused streptozotocin-induced diabetic rats
.
Naunyn Schmiedebergs Arch Pharmacol
1998
;
358
:
529
–
537
.
145Tesfamariam
B
, Cohen
RA.
Free radicals mediate endothelial cell dysfunction caused by elevated glucose
.
Am J Physiol
1992
;
263
:
H321
–
326
.
146Wong
ND
, Rozanski
A
, Gransar
H
, Miranda-Peats
R
, Kang
XP
, Hayes
S
, Shaw
L
, Friedman
J
, Polk
D
, Berman
DS.
Metabolic syndrome and diabetes are associated with an increased likelihood of inducible myocardial ischemia among patients with subclinical atherosclerosis
.
Diabetes Care
2005
;
28
:
1445
–
1450
.
147Goyal
BR
, Mehta
AA.
Diabetic cardiomyopathy: pathophysiological mechanisms and cardiac dysfuntion
.
Hum Exp Toxicol
2013
;
32
:
571
–
590
.
148Bornfeldt
KE
, Tabas
I.
Insulin resistance, hyperglycemia, and atherosclerosis
.
Cell Metab
2011
;
14
:
575
–
585
.
149Ingelsson
E
, Sundstrom
J
, Arnlov
J
, Zethelius
B
, Lind
L.
Insulin resistance and risk of congestive heart failure
.
JAMA
2005
;
294
:
334
–
341
.
150Arnlov
J
, Lind
L
, Zethelius
B
, Andren
B
, Hales
CN
, Vessby
B
, Lithell
H.
Several factors associated with the insulin resistance syndrome are predictors of left ventricular systolic dysfunction in a male population after 20 years of follow-up
.
Am Heart J
2001
;
142
:
720
–
724
.
151Karwi
QG
, Jorg
AR
, Lopaschuk
GD.
Allosteric, transcriptional and post-translational control of mitochondrial energy metabolism
.
Biochem J
2019
;
476
:
1695
–
1712
.
152Cook
SA
, Varela-Carver
A
, Mongillo
M
, Kleinert
C
, Khan
MT
, Leccisotti
L
, Strickland
N
, Matsui
T
, Das
S
, Rosenzweig
A
, Punjabi
P
, Camici
PG.
Abnormal myocardial insulin signalling in type 2 diabetes and left-ventricular dysfunction
.
Eur Heart J
2010
;
31
:
100
–
111
.
153Jia
G
, DeMarco
VG
, Sowers
JR.
Insulin resistance and hyperinsulinaemia in diabetic cardiomyopathy
.
Nat Rev Endocrinol
2016
;
12
:
144
–
153
.
154Battiprolu
PK
, Hojayev
B
, Jiang
N
, Wang
ZV
, Luo
X
, Iglewski
M
, Shelton
JM
, Gerard
RD
, Rothermel
BA
, Gillette
TG
, Lavandero
S
, Hill
JA.
Metabolic stress-induced activation of FoxO1 triggers diabetic cardiomyopathy in mice
.
J Clin Invest
2012
;
122
:
1109
–
1118
.
155Song
R
, Peng
W
, Zhang
Y
, Lv
F
, Wu
HK
, Guo
J
, Cao
Y
, Pi
Y
, Zhang
X
, Jin
L
, Zhang
M
, Jiang
P
, Liu
F
, Meng
S
, Zhang
X
, Jiang
P
, Cao
CM
, Xiao
RP.
Central role of E3 ubiquitin ligase MG53 in insulin resistance and metabolic disorders
.
Nature
2013
;
494
:
375
–
379
.
156Liu
F
, Song
R
, Feng
Y
, Guo
J
, Chen
Y
, Zhang
Y
, Chen
T
, Wang
Y
, Huang
Y
, Li
CY
, Cao
C
, Zhang
Y
, Hu
X
, Xiao
RP.
Upregulation of MG53 induces diabetic cardiomyopathy through transcriptional activation of peroxisome proliferation-activated receptor alpha
.
Circulation
2015
;
131
:
795
–
804
.
157Zhang
L
, Ussher
JR
, Oka
T
, Cadete
VJ
, Wagg
C
, Lopaschuk
GD.
Cardiac diacylglycerol accumulation in high fat-fed mice is associated with impaired insulin-stimulated glucose oxidation
.
Cardiovasc Res
2011
;
89
:
148
–
156
.
158Ussher
JR
, Koves
TR
, Cadete
VJ
, Zhang
L
, Jaswal
JS
, Swyrd
SJ
, Lopaschuk
DG
, Proctor
SD
, Keung
W
, Muoio
DM
, Lopaschuk
GD.
Inhibition of de novo ceramide synthesis reverses diet-induced insulin resistance and enhances whole-body oxygen consumption
.
Diabetes
2010
;
59
:
2453
–
2464
.
159Ussher
JR
, Folmes
CD
, Keung
W
, Fillmore
N
, Jaswal
JS
, Cadete
VJ
, Beker
DL
, Lam
VH
, Zhang
L
, Lopaschuk
GD.
Inhibition of serine palmitoyl transferase I reduces cardiac ceramide levels and increases glycolysis rates following diet-induced insulin resistance
.
PLoS One
2012
;
7
:
e37703
.
160Lopaschuk
GD.
Metabolic modulators in heart disease: past, present, and future
.
Can J Cardiol
2017
;
33
:
838
–
849
.
161Karwi
QG
, Uddin
GM
, Ho
KL
, Lopaschuk
GD.
Loss of Metabolic Flexibility in the Failing Heart
.
Front Cardiovasc Med
2018
;
5
:
1
–19.
162Greenwell
AA
, Gopal
K
, Ussher
JR.
Myocardial Energy Metabolism in Non-ischemic Cardiomyopathy
.
Front Physiol
2020
;
11
:
570421
–570438.
163Ussher
JR
, Lopaschuk
GD.
Cardiac insulin resistance: it's sweeter than you think
.
Endocrinology
2013
;
154
:
2575
–
2578
.
164Lopaschuk
GD
, Russell
JC.
Myocardial function and energy substrate metabolism in the insulin-resistant JCR: LA corpulent rat
.
J Appl Physiol (1985)
1991
;
71
:
1302
–
1308
.
165Kota
SK
, Kota
SK
, Jammula
S
, Panda
S
, Modi
KD.
Effect of diabetes on alteration of metabolism in cardiac myocytes: therapeutic implications
.
Diabetes Technol Ther
2011
;
13
:
1155
–
1160
.
166Karwi
Q
, Lopaschuk
GD.
Energy metabolism in patients with diabetes and heart failure
.
Heart Metab
2019
;
32
–
36
.
167How
OJ
, Aasum
E
, Severson
DL
, Chan
WY
, Essop
MF
, Larsen
TS.
Increased myocardial oxygen consumption reduces cardiac efficiency in diabetic mice
.
Diabetes
2006
;
55
:
466
–
473
.
168Ouwens
DM
, Diamant
M
, Fodor
M
, Habets
DDJ
, Pelsers
M
, El Hasnaoui
M
, Dang
ZC
, van den Brom
CE
, Vlasblom
R
, Rietdijk
A
, Boer
C
, Coort
SLM
, Glatz
JFC
, Luiken
J.
Cardiac contractile dysfunction in insulin-resistant rats fed a high-fat diet is associated with elevated CD36-mediated fatty acid uptake and esterification
.
Diabetologia
2007
;
50
:
1938
–
1948
.
169Sambandam
N
, Abrahani
MA
, St Pierre
E
, Al-Atar
O
, Cam
MC
, Rodrigues
B.
Localization of lipoprotein lipase in the diabetic heart: regulation by acute changes in insulin
.
Arterioscler Thromb Vasc Biol
1999
;
19
:
1526
–
1534
.
170Luiken
JJ
, Turcotte
LP
, Bonen
A.
Protein-mediated palmitate uptake and expression of fatty acid transport proteins in heart giant vesicles
.
J Lipid Res
1999
;
40
:
1007
–
1016
.
171Luiken
JJ
, Arumugam
Y
, Bell
RC
, Calles-Escandon
J
, Tandon
NN
, Glatz
JF
, Bonen
A.
Changes in fatty acid transport and transporters are related to the severity of insulin deficiency
.
Am J Physiol Endocrinol Metab
2002
;
283
:
E612
–
621
.
172Carley
AN
, Atkinson
LL
, Bonen
A
, Harper
ME
, Kunnathu
S
, Lopaschuk
GD
, Severson
DL.
Mechanisms responsible for enhanced fatty acid utilization by perfused hearts from type 2 diabetic db/db mice
.
Arch Physiol Biochem
2007
;
113
:
65
–
75
.
173Kim
TT
, Dyck
JR.
The role of CD36 in the regulation of myocardial lipid metabolism
.
Biochim Biophys Acta
2016
;
1861
:
1450
–
1460
.
174An
D
, Rodrigues
B.
Role of changes in cardiac metabolism in development of diabetic cardiomyopathy
.
Am J Physiol Heart Circ Physiol
2006
;
291
:
H1489
–
1506
.
175Varma
U
, Koutsifeli
P
, Benson
VL
, Mellor
KM
, Delbridge
LMD.
Molecular mechanisms of cardiac pathology in diabetes—experimental insights
.
Biochim Biophys Acta Mol Basis Dis
2018
;
1864
:
1949
–
1959
.
176Finck
BN
, Han
X
, Courtois
M
, Aimond
F
, Nerbonne
JM
, Kovacs
A
, Gross
RW
, Kelly
DP.
A critical role for PPARalpha-mediated lipotoxicity in the pathogenesis of diabetic cardiomyopathy: modulation by dietary fat content
.
Proc Natl Acad Sci USA
2003
;
100
:
1226
–
1231
.
177Kyriazis
ID
, Hoffman
M
, Gaignebet
L
, Lucchese
AM
, Markopoulou
E
, Palioura
D
, Wang
C
, Bannister
TD
, Christofidou-Solomidou
M
, Oka
SI
, Sadoshima
J
, Koch
WJ
, Goldberg
IJ
, Yang
VW
, Bialkowska
AB
, Kararigas
G
, Drosatos
K.
KLF5 is induced by FOXO1 and causes oxidative stress and diabetic cardiomyopathy
.
Circ Res
2021
;
128
:
335
–
357
.
178Kalliora
C
, Kyriazis
ID
, Oka
SI
, Lieu
MJ
, Yue
Y
, Area-Gomez
E
, Pol
CJ
, Tian
Y
, Mizushima
W
, Chin
A
, Scerbo
D
, Schulze
PC
, Civelek
M
, Sadoshima
J
, Madesh
M
, Goldberg
IJ
, Drosatos
K.
Dual peroxisome-proliferator-activated-receptor-alpha/gamma activation inhibits SIRT1-PGC1alpha axis and causes cardiac dysfunction
.
JCI Insight
2019
;
4
.
179Son
NH
, Yu
S
, Tuinei
J
, Arai
K
, Hamai
H
, Homma
S
, Shulman
GI
, Abel
ED
, Goldberg
IJ.
PPARgamma-induced cardiolipotoxicity in mice is ameliorated by PPARalpha deficiency despite increases in fatty acid oxidation
.
J Clin Invest
2010
;
120
:
3443
–
3454
.
180Song
S
, Attia
RR
, Connaughton
S
, Niesen
MI
, Ness
GC
, Elam
MB
, Hori
RT
, Cook
GA
, Park
EA.
Peroxisome proliferator activated receptor alpha (PPARalpha) and PPAR gamma coactivator (PGC-1alpha) induce carnitine palmitoyltransferase IA (CPT-1A) via independent gene elements
.
Mol Cell Endocrinol
2010
;
325
:
54
–
63
.
181Rasmussen
BB
, Holmback
UC
, Volpi
E
, Morio-Liondore
B
, Paddon-Jones
D
, Wolfe
RR.
Malonyl coenzyme A and the regulation of functional carnitine palmitoyltransferase-1 activity and fat oxidation in human skeletal muscle
.
J Clin Invest
2002
;
110
:
1687
–
1693
.
182Sakamoto
J
, Barr
RL
, Kavanagh
KM
, Lopaschuk
GD.
Contribution of malonyl-CoA decarboxylase to the high fatty acid oxidation rates seen in the diabetic heart
.
Am J Physiol Heart Circ Physiol
2000
;
278
:
H1196
–
H1204
.
183Saddik
M
, Gamble
J
, Witters
LA
, Lopaschuk
GD.
Acetyl-CoA carboxylase regulation of fatty acid oxidation in the heart
.
J Biol Chem
1993
;
268
:
25836
–
25845
.
184Dyck
JR
, Lopaschuk
GD.
Glucose metabolism, H+ production and Na+/H+-exchanger mRNA levels in ischemic hearts from diabetic rats. In Abdel-Aleem S, Lowe JE (eds).
Cardiac Metabolism in Health and Disease
.
Springer
,
1998
. pp.
85
–
93
.
185Wende
AR.
Post-translational modifications of the cardiac proteome in diabetes and heart failure
.
Proteomics Clin Appl
2016
;
10
:
25
–
38
.
186Fukushima
A
, Lopaschuk
GD.
Acetylation control of cardiac fatty acid beta-oxidation and energy metabolism in obesity, diabetes, and heart failure
.
Biochim Biophys Acta
2016
;
1862
:
2211
–
2220
.
187Vadvalkar
SS
, Baily
CN
, Matsuzaki
S
, West
M
, Tesiram
YA
, Humphries
KM.
Metabolic inflexibility and protein lysine acetylation in heart mitochondria of a chronic model of type 1 diabetes
.
Biochem J
2013
;
449
:
253
–
261
.
188Sun
Y
, Tian
Z
, Liu
N
, Zhang
L
, Gao
Z
, Sun
X
, Yu
M
, Wu
J
, Yang
F
, Zhao
Y
, Ren
H
, Chen
H
, Zhao
D
, Wang
Y
, Dong
S
, Xu
C
, Lu
F
, Zhang
W.
Exogenous H2S switches cardiac energy substrate metabolism by regulating SIRT3 expression in db/db mice
.
J Mol Med (Berl)
2018
;
96
:
281
–
299
.
189Saddik
M
, Lopaschuk
GD.
Triacylglycerol turnover in isolated working hearts of acutely diabetic rats
.
Can J Physiol Pharmacol
1994
;
72
:
1110
–
1119
.
190Borisov
AB
, Ushakov
AV
, Zagorulko
AK
, Novikov
NY
, Selivanova
KF
, Edwards
CA
, Russell
MW.
Intracardiac lipid accumulation, lipoatrophy of muscle cells and expansion of myocardial infarction in type 2 diabetic patients
.
Micron
2008
;
39
:
944
–
951
.
191O’Donnell
JM
, Zampino
M
, Alpert
NM
, Fasano
MJ
, Geenen
DL
, Lewandowski
ED.
Accelerated triacylglycerol turnover kinetics in hearts of diabetic rats include evidence for compartmented lipid storage
.
Am J Physiol Endocrinol Metab
2006
;
290
:
E448
–
455
.
192Paulson
DJ
, Crass
MF
3rd.
Endogenous triacylglycerol metabolism in diabetic heart
.
Am J Physiol
1982
;
242
:
H1084
–
1094
.
193Larsen
TS
, Severson
DL.
Influence of exogenous fatty acids and ketone bodies on rates of lipolysis in isolated ventricular myocytes from normal and diabetic rats
.
Can J Physiol Pharmacol
1990
;
68
:
1177
–
1182
.
194Chauhan
UP
, Singh
VN.
Myocardial phospholipid metabolism in alloxan diabetic rats
.
Life Sci
1978
;
22
:
1771
–
1776
.
195Lim
HY
, Wang
W
, Wessells
RJ
, Ocorr
K
, Bodmer
R.
Phospholipid homeostasis regulates lipid metabolism and cardiac function through SREBP signaling in Drosophila
.
Genes Dev
2011
;
25
:
189
–
200
.
196Ovide-Bordeaux
S
, Bescond-Jacquet
A
, Grynberg
A.
Cardiac mitochondrial alterations induced by insulin deficiency and hyperinsulinaemia in rats: targeting membrane homeostasis with trimetazidine
.
Clin Exp Pharmacol Physiol
2005
;
32
:
1061
–
1070
.
197Wall
SR
, Lopaschuk
GD.
Glucose oxidation rates in fatty acid-perfused isolated working hearts from diabetic rats
.
Biochim Biophys Acta
1989
;
1006
:
97
–
103
.
198Nicholl
TA
, Lopaschuk
GD
, McNeill
JH.
Effects of free fatty acids and dichloroacetate on isolated working diabetic rat heart
.
Am J Physiol
1991
;
261
:
H1053
–
1059
.
199Garvey
WT
, Hardin
D
, Juhaszova
M
, Dominguez
JH.
Effects of diabetes on myocardial glucose transport system in rats: implications for diabetic cardiomyopathy
.
Am J Physiol
1993
;
264
:
H837
–
844
.
200Hu
L
, Qiu
C
, Wang
X
, Xu
M
, Shao
X
, Wang
Y.
The association between diabetes mellitus and reduction in myocardial glucose uptake: a population-based (18)F-FDG PET/CT study
.
BMC Cardiovasc Disord
2018
;
18
:
203
.
201Camps
M
, Castello
A
, Munoz
P
, Monfar
M
, Testar
X
, Palacin
M
, Zorzano
A.
Effect of diabetes and fasting on GLUT-4 (muscle/fat) glucose-transporter expression in insulin-sensitive tissues. Heterogeneous response in heart, red and white muscle
.
Biochem J
1992
;
282(pt. 3
):
765
–
772
.
202Laughlin
MR
, Petit
WA
Jr, Shulman
RG
, Barrett
EJ.
Measurement of myocardial glycogen synthesis in diabetic and fasted rats
.
Am J Physiol
1990
;
258
:
E184
–
190
.
203Das
I.
Studies on glycogen metabolism in normal and diabetic rat heart in vivo
.
Can J Biochem
1973
;
51
:
637
–
641
.
204Nakao
M
, Matsubara
T
, Sakamoto
N.
Effects of diabetes on cardiac glycogen metabolism in rats
.
Heart Vessels
1993
;
8
:
171
–
175
.
205Yu
C
, Chen
Y
, Cline
GW
, Zhang
D
, Zong
H
, Wang
Y
, Bergeron
R
, Kim
JK
, Cushman
SW
, Cooney
GJ
, Atcheson
B
, White
MF
, Kraegen
EW
, Shulman
GI.
Mechanism by which fatty acids inhibit insulin activation of insulin receptor substrate-1 (IRS-1)-associated phosphatidylinositol 3-kinase activity in muscle
.
J Biol Chem
2002
;
277
:
50230
–
50236
.
206Ellis
BA
, Poynten
A
, Lowy
AJ
, Furler
SM
, Chisholm
DJ
, Kraegen
EW
, Cooney
GJ.
Long-chain acyl-CoA esters as indicators of lipid metabolism and insulin sensitivity in rat and human muscle
.
Am J Physiol Endocrinol Metab
2000
;
279
:
E554
–
560
.
207Bockus
LB
, Matsuzaki
S
, Vadvalkar
SS
, Young
ZT
, Giorgione
JR
, Newhardt
MF
, Kinter
M
, Humphries
KM.
Cardiac insulin signaling regulates glycolysis through phosphofructokinase 2 content and activity
.
J Am Heart Assoc
2017
;
6
:
e007159
.
208Da Silva
D
, Ausina
P
, Alencar
EM
, Coelho
WS
, Zancan
P
, Sola-Penna
M.
Metformin reverses hexokinase and phosphofructokinase downregulation and intracellular distribution in the heart of diabetic mice
.
IUBMB Life
2012
;
64
:
766
–
774
.
209Randle
PJ
, Garland
PB
, Hales
CN
, Newsholme
EA.
The glucose fatty-acid cycle. Its role in insulin sensitivity and the metabolic disturbances of diabetes mellitus
.
Lancet
1963
;
281
:
785
–
789
.
210Dewald
O
, Sharma
S
, Adrogue
J
, Salazar
R
, Duerr
GD
, Crapo
JD
, Entman
ML
, Taegtmeyer
H.
Downregulation of peroxisome proliferator-activated receptor-alpha gene expression in a mouse model of ischemic cardiomyopathy is dependent on reactive oxygen species and prevents lipotoxicity
.
Circulation
2005
;
112
:
407
–
415
.
211Mather
KJ
, Hutchins
GD
, Perry
K
, Territo
W
, Chisholm
R
, Acton
A
, Glick-Wilson
B
, Considine
RV
, Moberly
S
, DeGrado
TR.
Assessment of myocardial metabolic flexibility and work efficiency in human type 2 diabetes using 16-[18F]fluoro-4-thiapalmitate, a novel PET fatty acid tracer
.
Am J Physiol Endocrinol Metab
2016
;
310
:
E452
–
E460
.
212Rider
OJ
, Apps
A
, Miller
J
, Lau
JYC
, Lewis
AJM
, Peterzan
MA
, Dodd
MS
, Lau
AZ
, Trumper
C
, Gallagher
FA
, Grist
JT
, Brindle
KM
, Neubauer
S
, Tyler
DJ.
Noninvasive in vivo assessment of cardiac metabolism in the healthy and diabetic human heart using hyperpolarized 13C MRI
.
Circ Res
2020
;
126
:
725
–
736
.
213Rijzewijk
LJ
, van der Meer
RW
, Lamb
HJ
, de Jong
HW
, Lubberink
M
, Romijn
JA
, Bax
JJ
, de Roos
A
, Twisk
JW
, Heine
RJ
, Lammertsma
AA
, Smit
JW
, Diamant
M.
Altered myocardial substrate metabolism and decreased diastolic function in nonischemic human diabetic cardiomyopathy: studies with cardiac positron emission tomography and magnetic resonance imaging
.
J Am Coll Cardiol
2009
;
54
:
1524
–
1532
.
214Doria
A
, Nosadini
R
, Avogaro
A
, Fioretto
P
, Crepaldi
G.
Myocardial metabolism in type 1 diabetic patients without coronary artery disease
.
Diabet Med
1991
;
8 Spec No
:
S104
–
107
.
215Scheuermann-Freestone
M
, Madsen
PL
, Manners
D
, Blamire
AM
, Buckingham
RE
, Styles
P
, Radda
GK
, Neubauer
S
, Clarke
K.
Abnormal cardiac and skeletal muscle energy metabolism in patients with type 2 diabetes
.
Circulation
2003
;
107
:
3040
–
3046
.
216Neubauer
S
, Horn
M
, Cramer
M
, Harre
K
, Newell
JB
, Peters
W
, Pabst
T
, Ertl
G
, Hahn
D
, Ingwall
JS
, Kochsiek
K.
Myocardial phosphocreatine-to-ATP ratio is a predictor of mortality in patients with dilated cardiomyopathy
.
Circulation
1997
;
96
:
2190
–
2196
.
217Ritchie
RH
, Abel
ED.
Basic mechanisms of diabetic heart disease
.
Circ Res
2020
;
126
:
1501
–
1525
.
218Mereweather
LJ
, Montes Aparicio
CN
, Heather
LC.
Positioning metabolism as a central player in the diabetic heart
.
J Lipid Atheroscler
2020
;
9
:
92
–
109
.
219van den Brom
CE
, Huisman
MC
, Vlasblom
R
, Boontje
NM
, Duijst
S
, Lubberink
M
, Molthoff
CF
, Lammertsma
AA
, van der Velden
J
, Boer
C
, Ouwens
DM
, Diamant
M.
Altered myocardial substrate metabolism is associated with myocardial dysfunction in early diabetic cardiomyopathy in rats: studies using positron emission tomography
.
Cardiovasc Diabetol
2009
;
8
:
39
.
220Lopaschuk
GD
, Spafford
M.
Response of isolated working hearts to fatty acids and carnitine palmitoyltransferase I inhibition during reduction of coronary flow in acutely and chronically diabetic rats
.
Circ Res
1989
;
65
:
378
–
387
.
221Lopaschuk
GD
, Saddik
M
, Barr
R
, Huang
L
, Barker
CC
, Muzyka
RA.
Effects of high levels of fatty acids on functional recovery of ischemic hearts from diabetic rats
.
Am J Physiol Endocrinol Metab
2006
;
263
:
E1046
–
E1053
.
222Karwi
QG
, Zhang
L
, Altamimi
TR
, Wagg
CS
, Patel
V
, Uddin
GM
, Joerg
AR
, Padwal
RS
, Johnstone
DE
, Sharma
A
, Oudit
GY
, Lopaschuk
GD.
Weight loss enhances cardiac energy metabolism and function in heart failure associated with obesity
.
Diabetes Obes Metab
2019
;
21
:
1944
–
1955
.
223Sankaralingam
S
, Abo Alrob
O
, Zhang
L
, Jaswal
JS
, Wagg
CS
, Fukushima
A
, Padwal
RS
, Johnstone
DE
, Sharma
AM
, Lopaschuk
GD.
Lowering body weight in obese mice with diastolic heart failure improves cardiac insulin sensitivity and function: implications for the obesity paradox
.
Diabetes
2015
;
64
:
1643
–
1657
.
224Sun
H
, Olson
KC
, Gao
C
, Prosdocimo
DA
, Zhou
M
, Wang
Z
, Jeyaraj
D
, Youn
JY
, Ren
S
, Liu
Y
, Rau
CD
, Shah
S
, Ilkayeva
O
, Gui
WJ
, William
NS
, Wynn
RM
, Newgard
CB
, Cai
H
, Xiao
X
, Chuang
DT
, Schulze
PC
, Lynch
C
, Jain
MK
, Wang
Y.
Catabolic defect of branched-chain amino acids promotes heart failure
.
Circulation
2016
;
133
:
2038
–
2049
.
225Du
X
, Li
Y
, Wang
Y
, You
H
, Hui
P
, Zheng
Y
, Du
J.
Increased branched-chain amino acid levels are associated with long-term adverse cardiovascular events in patients with STEMI and acute heart failure
.
Life Sci
2018
;
209
:
167
–
172
.
226Lopaschuk
GD
, Karwi
QG
, Ho
KL
, Pherwani
S
, Ketema
EB.
Ketone metabolism in the failing heart
.
Biochim Biophys Acta Mol Cell Biol Lipids
2020
;
1865
:
158813
.
227Voros
G
, Ector
J
, Garweg
C
, Droogne
W
, Van Cleemput
J
, Peersman
N
, Vermeersch
P
, Janssens
S.
Increased cardiac uptake of ketone bodies and free fatty acids in human heart failure and hypertrophic left ventricular remodeling
.
Circ Heart Fail
2018
;
11
:
e004953
.
228Lopaschuk
GD
, Verma
S.
Mechanisms of cardiovascular benefits of sodium glucose co-transporter 2 (SGLT2) inhibitors: a state-of-the-art review
.
JACC Basic Transl Sci
2020
;
5
:
632
–
644
.
229Brahma
MK
, Ha
CM
, Pepin
ME
, Mia
S
, Sun
Z
, Chatham
JC
, Habegger
KM
, Abel
ED
, Paterson
AJ
, Young Me
WA.
Increased glucose availability attenuates myocardial ketone body utilization
.
J Am Heart Assoc
2020
;
9
:
e013039
.
230Fillmore
N
, Wagg
CS
, Zhang
LY
, Fukushima
A
, Lopaschuk
GD.
Cardiac branched-chain amino acid oxidation is reduced during insulin resistance in the heart
.
Am J Physiol Endocrinol Metab
2018
;
315
:
E1046
–
E1052
.
231Uddin
GM
, Zhang
L
, Shah
S
, Fukushima
A
, Wagg
CS
, Gopal
K
, Al Batran
R
, Pherwani
S
, Ho
KL
, Boisvenue
J
, Karwi
QG
, Altamimi
T
, Wishart
DS
, Dyck
JRB
, Ussher
JR
, Oudit
GY
, Lopaschuk
GD.
Impaired branched chain amino acid oxidation contributes to cardiac insulin resistance in heart failure
.
Cardiovasc Diabetol
2019
;
18
:
86
.
232Karwi
QG
, Zhang
L
, Wagg
CS
, Wang
W
, Ghandi
M
, Thai
D
, Yan
H
, Ussher
JR
, Oudit
GY
, Lopaschuk
GD.
Targeting the glucagon receptor improves cardiac function and enhances insulin sensitivity following a myocardial infarction
.
Cardiovasc Diabetol
2019
;
18
:
1
–18.
233Marfella
R
, Di Filippo
C
, Portoghese
M
, Barbieri
M
, Ferraraccio
F
, Siniscalchi
M
, Cacciapuoti
F
, Rossi
F
, D'Amico
M
, Paolisso
G.
Myocardial lipid accumulation in patients with pressure-overloaded heart and metabolic syndrome
.
J Lipid Res
2009
;
50
:
2314
–
2323
.
234McGavock
JM
, Lingvay
I
, Zib
I
, Tillery
T
, Salas
N
, Unger
R
, Levine
BD
, Raskin
P
, Victor
RG
, Szczepaniak
LS.
Cardiac steatosis in diabetes mellitus: a 1H-magnetic resonance spectroscopy study
.
Circulation
2007
;
116
:
1170
–
1175
.
235Barchuk
M
, Schreier
L
, Lopez
G
, Cevey
A
, Baldi
J
, Fernandez Tome
MDC
, Goren
N
, Rubio
M
, Miksztowicz
V
, Berg
G.
Glycosylphosphatidylinositol-anchored high density lipoprotein-binding protein 1 and angiopoietin-like protein 4 are associated with the increase of lipoprotein lipase activity in epicardial adipose tissue from diabetic patients
.
Atherosclerosis
2019
;
288
:
51
–
59
.
236Yang
J
, Sambandam
N
, Han
X
, Gross
RW
, Courtois
M
, Kovacs
A
, Febbraio
M
, Finck
BN
, Kelly
DP.
CD36 deficiency rescues lipotoxic cardiomyopathy
.
Circ Res
2007
;
100
:
1208
–
1217
.
237Duncan
JG
, Bharadwaj
KG
, Fong
JL
, Mitra
R
, Sambandam
N
, Courtois
MR
, Lavine
KJ
, Goldberg
IJ
, Kelly
DP.
Rescue of cardiomyopathy in peroxisome proliferator-activated receptor-alpha transgenic mice by deletion of lipoprotein lipase identifies sources of cardiac lipids and peroxisome proliferator-activated receptor-alpha activators
.
Circulation
2010
;
121
:
426
–
435
.
238Bastie
CC
, Hajri
T
, Drover
VA
, Grimaldi
PA
, Abumrad
NA.
CD36 in myocytes channels fatty acids to a lipase-accessible triglyceride pool that is related to cell lipid and insulin responsiveness
.
Diabetes
2004
;
53
:
2209
–
2216
.
239Febbraio
M
, Guy
E
, Coburn
C
, Knapp
FF
, Beets
AL
, Abumrad
NA
, Silverstein
RL.
The impact of overexpression and deficiency of fatty acid translocase (FAT)/CD36
.
Mol Cell Biochem
2002
;
239
:
193
–
197
.
240Chiu
HC
, Kovacs
A
, Blanton
RM
, Han
X
, Courtois
M
, Weinheimer
CJ
, Yamada
KA
, Brunet
S
, Xu
H
, Nerbonne
JM
, Welch
MJ
, Fettig
NM
, Sharp
TL
, Sambandam
N
, Olson
KM
, Ory
DS
, Schaffer
JE.
Transgenic expression of fatty acid transport protein 1 in the heart causes lipotoxic cardiomyopathy
.
Circ Res
2005
;
96
:
225
–
233
.
241Zlobine
I
, Gopal
K
, Ussher
JR.
Lipotoxicity in obesity and diabetes-related cardiac dysfunction
.
Biochim Biophys Acta
2016
;
1861
:
1555
–
1568
.
242Takada
I
, Makishima
M.
Peroxisome proliferator-activated receptor agonists and antagonists: a patent review (2014-present)
.
Expert Opin Ther Pat
2020
;
30
:
1
–
13
.
243Dyck
JR
, Hopkins
TA
, Bonnet
S
, Michelakis
ED
, Young
ME
, Watanabe
M
, Kawase
Y
, Jishage
K
, Lopaschuk
GD.
Absence of malonyl coenzyme A decarboxylase in mice increases cardiac glucose oxidation and protects the heart from ischemic injury
.
Circ
2006
;
114
:
1721
–
1728
.
244Goldberg
IJ
, Trent
CM
, Schulze
PC.
Lipid metabolism and toxicity in the heart
.
Cell Metab
2012
;
15
:
805
–
812
.
245Park
TS
, Hu
YY
, Noh
HL
, Drosatos
K
, Okajima
K
, Buchanan
J
, Tuinei
J
, Homma
S
, Jiang
XC
, Abel
ED
, Goldberg
IJ.
Ceramide is a cardiotoxin in lipotoxic cardiomyopathy
.
J Lipid Res
2008
;
49
:
2101
–
2112
.
246Inoguchi
T
, Battan
R
, Handler
E
, Sportsman
JR
, Heath
W
, King
GL.
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 USA
1992
;
89
:
11059
–
11063
.
247Wang
G
, Silva
J
, Krishnamurthy
K
, Tran
E
, Condie
BG
, Bieberich
E.
Direct binding to ceramide activates protein kinase Czeta before the formation of a pro-apoptotic complex with PAR-4 in differentiating stem cells
.
J Biol Chem
2005
;
280
:
26415
–
26424
.
248Chuang
TT
, LeVine
H
3rd, De Blasi
A.
Phosphorylation and activation of beta-adrenergic receptor kinase by protein kinase C
.
J Biol Chem
1995
;
270
:
18660
–
18665
.
249Drosatos
K
, Bharadwaj
KG
, Lymperopoulos
A
, Ikeda
S
, Khan
R
, Hu
Y
, Agarwal
R
, Yu
S
, Jiang
H
, Steinberg
SF
, Blaner
WS
, Koch
WJ
, Goldberg
IJ.
Cardiomyocyte lipids impair beta-adrenergic receptor function via PKC activation
.
Am J Physiol Endocrinol Metab
2011
;
300
:
E489
–
499
.
250Koch
WJ
, Rockman
HA
, Samama
P
, Hamilton
RA
, Bond
RA
, Milano
CA
, Lefkowitz
RJ.
Cardiac function in mice overexpressing the beta-adrenergic receptor kinase or a beta ARK inhibitor
.
Science
1995
;
268
:
1350
–
1353
.
251Itani
SI
, Zhou
Q
, Pories
WJ
, MacDonald
KG
, Dohm
GL.
Involvement of protein kinase C in human skeletal muscle insulin resistance and obesity
.
Diabetes
2000
;
49
:
1353
–
1358
.
252Li
Y
, Soos
TJ
, Li
X
, Wu
J
, Degennaro
M
, Sun
X
, Littman
DR
, Birnbaum
MJ
, Polakiewicz
RD.
Protein kinase C Theta inhibits insulin signaling by phosphorylating IRS1 at Ser(1101)
.
J Biol Chem
2004
;
279
:
45304
–
45307
.
253Suzuki
J
, Ueno
M
, Uno
M
, Hirose
Y
, Zenimaru
Y
, Takahashi
S
, Osuga
J
, Ishibashi
S
, Takahashi
M
, Hirose
M
, Yamada
M
, Kraemer
FB
, Miyamori
I.
Effects of hormone-sensitive lipase disruption on cardiac energy metabolism in response to fasting and refeeding
.
Am J Physiol Endocrinol Metab
2009
;
297
:
E1115
–
1124
.
254Listenberger
LL
, Han
X
, Lewis
SE
, Cases
S
, Farese
RV
Jr, Ory
DS
, Schaffer
JE.
Triglyceride accumulation protects against fatty acid-induced lipotoxicity
.
Proc Natl Acad Sci USA
2003
;
100
:
3077
–
3082
.
255Isfort
M
, Stevens
SC
, Schaffer
S
, Jong
CJ
, Wold
LE.
Metabolic dysfunction in diabetic cardiomyopathy
.
Heart Fail Rev
2014
;
19
:
35
–
48
.
256Verma
S
, Rawat
S
, Ho
KL
, Wagg
CS
, Zhang
LY
, Teoh
H
, Dyck
JE
, Uddin
GM
, Oudit
GY
, Mayoux
E
, Lehrke
M
, Marx
N
, Lopaschuk
GD.
Empagliflozin increases cardiac energy production in diabetes novel translational insights into the heart failure benefits of SGLT2 inhibitors
.
JACC Basic Trans Sci
2018
;
3
:
576
–
587
.
257Amaral
N
, Okonko
DO.
Metabolic abnormalities of the heart in type II diabetes
.
Diab Vasc Dis Res
2015
;
12
:
239
–
248
.
258Wende
AR
, Schell
JC
, Ha
CM
, Pepin
ME
, Khalimonchuk
O
, Schwertz
H
, Pereira
RO
, Brahma
MK
, Tuinei
J
, Contreras-Ferrat
A
, Wang
L
, Andrizzi
CA
, Olsen
CD
, Bradley
WE
, Dell'Italia
LJ
, Dillmann
WH
, Litwin
SE
, Abel
ED.
Maintaining myocardial glucose utilization in diabetic cardiomyopathy accelerates mitochondrial dysfunction
.
Diabetes
2020
;
69
:
2094
–
2111
.
259Bugger
H
, Pfeil
K.
Mitochondrial ROS in myocardial ischemia reperfusion and remodeling
.
Biochim Biophys Acta Mol Basis Dis
2020
;
1866
:
165768
.
260Song
M
, Chen
Y
, Gong
G
, Murphy
E
, Rabinovitch
PS
, Dorn
GW
2nd.
Super-suppression of mitochondrial reactive oxygen species signaling impairs compensatory autophagy in primary mitophagic cardiomyopathy
.
Circ Res
2014
;
115
:
348
–
353
.
261Fukushima
A
, Lopaschuk
GD.
Cardiac fatty acid oxidation in heart failure associated with obesity and diabetes
.
Biochim Biophys Acta
2016
;
1861
:
1525
–
1534
.
262Newsholme
P
, Haber
EP
, Hirabara
SM
, Rebelato
EL
, Procopio
J
, Morgan
D
, Oliveira-Emilio
HC
, Carpinelli
AR
, Curi
R.
Diabetes associated cell stress and dysfunction: role of mitochondrial and non-mitochondrial ROS production and activity
.
J Physiol
2007
;
583
:
9
–
24
.
263Boudina
S
, Sena
S
, Theobald
H
, Sheng
X
, Wright
JJ
, Hu
XX
, Aziz
S
, Johnson
JI
, Bugger
H
, Zaha
VG
, Abel
ED.
Mitochondrial energetics in the heart in obesity-related diabetes: direct evidence for increased uncoupled respiration and activation of uncoupling proteins
.
Diabetes
2007
;
56
:
2457
–
2466
.
264Nishikawa
T
, Edelstein
D
, Du
XL
, Yamagishi
S
, Matsumura
T
, Kaneda
Y
, Yorek
MA
, Beebe
D
, Oates
PJ
, Hammes
HP
, Giardino
I
, Brownlee
M.
Normalizing mitochondrial superoxide production blocks three pathways of hyperglycaemic damage
.
Nature
2000
;
404
:
787
–
790
.
265Teshima
Y
, Takahashi
N
, Nishio
S
, Saito
S
, Kondo
H
, Fukui
A
, Aoki
K
, Yufu
K
, Nakagawa
M
, Saikawa
T.
Production of reactive oxygen species in the diabetic heart. Roles of mitochondria and NADPH oxidase
.
Circ J
2014
;
78
:
300
–
306
.
266Wilson
AJ
, Gill
EK
, Abudalo
RA
, Edgar
KS
, Watson
CJ
, Grieve
DJ.
Reactive oxygen species signalling in the diabetic heart: emerging prospect for therapeutic targeting
.
Heart
2018
;
104
:
293
–
299
.
267Bugger
H
, Abel
ED.
Molecular mechanisms of diabetic cardiomyopathy
.
Diabetologia
2014
;
57
:
660
–
671
.
268Kaludercic
N
, Di Lisa
F.
Mitochondrial ROS formation in the pathogenesis of diabetic cardiomyopathy
.
Front Cardiovasc Med
2020
;
7
:
1
–15.
269Brookes
PS
, Yoon
Y
, Robotham
JL
, Anders
MW
, Sheu
SS.
Calcium, ATP, and ROS: a mitochondrial love-hate triangle
.
Am J Physiol Cell Physiol
2004
;
287
:
C817
–
833
.
270Yan
J
, Young
ME
, Cui
L
, Lopaschuk
GD
, Liao
R
, Tian
R.
Increased glucose uptake and oxidation in mouse hearts prevent high fatty acid oxidation but cause cardiac dysfunction in diet-induced obesity
.
Circulation
2009
;
119
:
2818
–
U2131
.
271Santos
CX
, Raza
S
, Shah
AM.
Redox signaling in the cardiomyocyte: from physiology to failure
.
Int J Biochem Cell Biol
2016
;
74
:
145
–
151
.
272Sharma
A
, Tate
M
, Mathew
G
, Vince
JE
, Ritchie
RH
, de Haan
JB.
Oxidative stress and NLRP3-inflammasome activity as significant drivers of diabetic cardiovascular complications: therapeutic implications
.
Front Physiol
2018
;
9
:
1
–15.
273Ge
Q
, Zhao
L
, Ren
XM
, Ye
P
, Hu
ZY.
LCZ696, an angiotensin receptor-neprilysin inhibitor, ameliorates diabetic cardiomyopathy by inhibiting inflammation, oxidative stress and apoptosis
.
Exp Biol Med (Maywood)
2019
;
244
:
1028
–
1039
.
274Luo
W
, Jin
Y
, Wu
G
, Zhu
W
, Qian
Y
, Zhang
Y
, Li
J
, Zhu
A
, Liang
G.
Blockage of ROS and MAPKs-mediated inflammation via restoring SIRT1 by a new compound LF10 prevents type 1 diabetic cardiomyopathy
.
Toxicol Appl Pharmacol
2019
;
370
:
24
–
35
.
275Eid
RA
, Alkhateeb
MA
, El-Kott
AF
, Eleawa
SM
, Zaki
MSA
, Alaboodi
SA
, Salem Al-Shudiefat
AA
, Aldera
H
, Alnamar
NM
, Alassiri
M
, Khalil
MA.
A high-fat diet rich in corn oil induces cardiac fibrosis in rats by activating JAK2/STAT3 and subsequent activation of ANG II/TGF-1beta/Smad3 pathway: the role of ROS and IL-6 trans-signaling
.
J Food Biochem
2019
;
43
:
e12952
.
276Yang
Y-C
, Tsai
C-Y
, Chen
C-L
, Kuo
C-H
, Hou
C-W
, Cheng
S-Y
, Aneja
R
, Huang
C-Y
, Kuo
W-W.
Pkcδ activation is involved in ROS-mediated mitochondrial dysfunction and apoptosis in cardiomyocytes exposed to advanced glycation end products (ages)
.
Aging Dis
2018
;
9
:
647
.
277Liu
Q
, Wang
S
, Cai
L.
Diabetic cardiomyopathy and its mechanisms: role of oxidative stress and damage
.
J Diabetes Invest
2014
;
5
:
623
–
634
.
278Ritchie
RH
, Love
JE
, Huynh
K
, Bernardo
BC
, Henstridge
DC
, Kiriazis
H
, Tham
YK
, Sapra
G
, Qin
C
, Cemerlang
N
, Boey
EJ
, Jandeleit-Dahm
K
, Du
XJ
, McMullen
JR.
Enhanced phosphoinositide 3-kinase(p110alpha) activity prevents diabetes-induced cardiomyopathy and superoxide generation in a mouse model of diabetes
.
Diabetologia
2012
;
55
:
3369
–
3381
.
279Joseph
LC
, Barca
E
, Subramanyam
P
, Komrowski
M
, Pajvani
U
, Colecraft
HM
, Hirano
M
, Morrow
JP.
Inhibition of NAPDH Oxidase 2 (NOX2) prevents oxidative stress and mitochondrial abnormalities caused by saturated fat in cardiomyocytes
.
PLoS One
2016
;
11
:
e0145750
.
280Wang
S
, Ding
L
, Ji
H
, Xu
Z
, Liu
Q
, Zheng
Y.
The role of p38 MAPK in the development of diabetic cardiomyopathy
.
Int J Mol Sci
2016
;
17
:
1037
.
281Huynh
K
, Bernardo
BC
, McMullen
JR
, Ritchie
RH.
Diabetic cardiomyopathy: mechanisms and new treatment strategies targeting antioxidant signaling pathways
.
Pharmacol Ther
2014
;
142
:
375
–
415
.
282Rajesh
M
, Mukhopadhyay
P
, Batkai
S
, Mukhopadhyay
B
, Patel
V
, Hasko
G
, Szabo
C
, Mabley
JG
, Liaudet
L
, Pacher
P.
Xanthine oxidase inhibitor allopurinol attenuates the development of diabetic cardiomyopathy
.
J Cell Mol Med
2009
;
13
:
2330
–
2341
.
283Huynh
K
, Kiriazis
H
, Du
XJ
, Love
JE
, Gray
SP
, Jandeleit-Dahm
KA
, McMullen
JR
, Ritchie
RH.
Targeting the upregulation of reactive oxygen species subsequent to hyperglycemia prevents type 1 diabetic cardiomyopathy in mice
.
Free Radic Biol Med
2013
;
60
:
307
–
317
.
284De Blasio
MJ
, Huynh
K
, Qin
C
, Rosli
S
, Kiriazis
H
, Ayer
A
, Cemerlang
N
, Stocker
R
, Du
XJ
, McMullen
JR
, Ritchie
RH.
Therapeutic targeting of oxidative stress with coenzyme Q10 counteracts exaggerated diabetic cardiomyopathy in a mouse model of diabetes with diminished PI3K(p110alpha) signaling
.
Free Radic Biol Med
2015
;
87
:
137
–
147
.
285Heart Outcomes Prevention Evaluation Study
I
, Yusuf
S
, Dagenais
G
, Pogue
J
, Bosch
J
, Sleight
P.
Vitamin E supplementation and cardiovascular events in high-risk patients
.
N Engl J Med
2000
;
342
:
154
–
160
.
286Sairam
T
, Patel
AN
, Subrahmanian
M
, Gopalan
R
, Pogwizd
SM
, Ramalingam
S
, Sankaran
R
, Rajasekaran
NS.
Evidence for a hyper-reductive redox in a sub-set of heart failure patients
.
J Transl Med
2018
;
16
:
130
.
287Ilkun
O
, Wilde
N
, Tuinei
J
, Pires
KM
, Zhu
Y
, Bugger
H
, Soto
J
, Wayment
B
, Olsen
C
, Litwin
SE
, Abel
ED.
Antioxidant treatment normalizes mitochondrial energetics and myocardial insulin sensitivity independently of changes in systemic metabolic homeostasis in a mouse model of the metabolic syndrome
.
J Mol Cell Cardiol
2015
;
85
:
104
–
116
.
288Dietl
A
, Maack
C.
Targeting mitochondrial calcium handling and reactive oxygen species in heart failure
.
Curr Heart Fail Rep
2017
;
14
:
338
–
349
.
289Boudina
S
, Bugger
H
, Sena
S
, O'Neill
BT
, Zaha
VG
, Ilkun
O
, Wright
JJ
, Mazumder
PK
, Palfreyman
E
, Tidwell
TJ
, Theobald
H
, Khalimonchuk
O
, Wayment
B
, Sheng
X
, Rodnick
KJ
, Centini
R
, Chen
D
, Litwin
SE
, Weimer
BE
, Abel
ED.
Contribution of impaired myocardial insulin signaling to mitochondrial dysfunction and oxidative stress in the heart
.
Circulation
2009
;
119
:
1272
–
1283
.
290Ma
W
, Guo
W
, Shang
F
, Li
Y
, Li
W
, Liu
J
, Ma
C
, Teng
J.
Bakuchiol alleviates hyperglycemia-induced diabetic cardiomyopathy by reducing myocardial oxidative stress via activating the SIRT1/Nrf2 signaling pathway
.
Oxid Med Cell Longev
2020
;
2020
:
3732718
.
291Dai
DF
, Chen
T
, Szeto
H
, Nieves-Cintron
M
, Kutyavin
V
, Santana
LF
, Rabinovitch
PS.
Mitochondrial targeted antioxidant peptide ameliorates hypertensive cardiomyopathy
.
J Am Coll Cardiol
2011
;
58
:
73
–
82
.
292Dai
DF
, Hsieh
EJ
, Chen
T
, Menendez
LG
, Basisty
NB
, Tsai
L
, Beyer
RP
, Crispin
DA
, Shulman
NJ
, Szeto
HH
, Tian
R
, MacCoss
MJ
, Rabinovitch
PS.
Global proteomics and pathway analysis of pressure-overload-induced heart failure and its attenuation by mitochondrial-targeted peptides
.
Circ Heart Fail
2013
;
6
:
1067
–
1076
.
293Karwi
QG
, Bornbaum
J
, Boengler
K
, Torregrossa
R
, Whiteman
M
, Wood
ME
, Schulz
R
, Baxter
GF.
AP39, a mitochondria-targeting hydrogen sulfide (H2 S) donor, protects against myocardial reperfusion injury independently of salvage kinase signalling
.
Br J Pharmacol
2017
;
174
:
287
–
301
.
294Gu
J
, Cheng
Y
, Wu
H
, Kong
L
, Wang
S
, Xu
Z
, Zhang
Z
, Tan
Y
, Keller
BB
, Zhou
H
, Wang
Y
, Xu
Z
, Cai
L.
Metallothionein is downstream of Nrf2 and partially mediates sulforaphane prevention of diabetic cardiomyopathy
.
Diabetes
2017
;
66
:
529
–
542
.
295Wang
JQ
, Wang
SD
, Wang
WN
, Chen
J
, Zhang
ZG
, Zheng
Q
, Liu
Q
, Cai
L.
Protection against diabetic cardiomyopathy is achieved using a combination of sulforaphane and zinc in type 1 diabetic OVE26 mice
.
J Cell Mol Med
2019
;
23
:
6319
–
6330
.
296Minamiyama
Y
, Bito
Y
, Takemura
S
, Takahashi
Y
, Kodai
S
, Mizuguchi
S
, Nishikawa
Y
, Suehiro
S
, Okada
S.
Calorie restriction improves cardiovascular risk factors via reduction of mitochondrial reactive oxygen species in type II diabetic rats
.
J Pharmacol Exp Ther
2007
;
320
:
535
–
543
.
297Tian
C
, Gao
L
, Zhang
A
, Hackfort
BT
, Zucker
IH.
Therapeutic effects of Nrf2 activation by bardoxolone methyl in chronic heart failure
.
J Pharmacol Exp Ther
2019
;
371
:
642
–
651
.
298Usui
M
, Matsuoka
H
, Miyazaki
H
, Ueda
S
, Okuda
S
, Imaizumi
T.
Increased endogenous nitric oxide synthase inhibitor in patients with congestive heart failure
.
Life Sci
1998
;
62
:
2425
–
2430
.
299Ge
ZD
, Lian
QQ
, Mao
XW
, Xia
ZY.
Current status and challenges of NRF2 as a potential therapeutic target for diabetic cardiomyopathy
.
Int Heart J
2019
;
60
:
512
–
520
.
300Yang
H
, Xu
W
, Zhou
Z
, Liu
J
, Li
X
, Chen
L
, Weng
J
, Yu
Z.
Curcumin attenuates urinary excretion of albumin in type II diabetic patients with enhancing nuclear factor erythroid-derived 2-like 2 (Nrf2) system and repressing inflammatory signaling efficacies
.
Exp Clin Endocrinol Diabetes
2015
;
123
:
360
–
367
.
301Cheng
D
, Li
W
, Wang
L
, Lin
T
, Poiani
G
, Wassef
A
, Hudlikar
R
, Ondar
P
, Brunetti
L
, Kong
AN.
Pharmacokinetics, pharmacodynamics, and PKPD modeling of curcumin in regulating antioxidant and epigenetic gene expression in healthy human volunteers
.
Mol Pharm
2019
;
16
:
1881
–
1889
.
302Bhaumik
S
, Jyothi
MD
, Khar
A.
Differential modulation of nitric oxide production by curcumin in host macrophages and NK cells
.
FEBS Lett
2000
;
483
:
78
–
82
.
303Jung
KK
, Lee
HS
, Cho
JY
, Shin
WC
, Rhee
MH
, Kim
TG
, Kang
JH
, Kim
SH
, Hong
S
, Kang
SY.
Inhibitory effect of curcumin on nitric oxide production from lipopolysaccharide-activated primary microglia
.
Life Sci
2006
;
79
:
2022
–
2031
.
304Kado
S
, Nagase
T
, Nagata
N.
Circulating levels of interleukin-6, its soluble receptor and interleukin-6/interleukin-6 receptor complexes in patients with type 2 diabetes mellitus
.
Acta Diabetol
1999
;
36
:
67
–
72
.
305Muller
S
, Martin
S
, Koenig
W
, Hanifi-Moghaddam
P
, Rathmann
W
, Haastert
B
, Giani
G
, Illig
T
, Thorand
B
, Kolb
H.
Impaired glucose tolerance is associated with increased serum concentrations of interleukin 6 and co-regulated acute-phase proteins but not TNF-alpha or its receptors
.
Diabetologia
2002
;
45
:
805
–
812
.
306Pickup
JC
, Chusney
GD
, Thomas
SM
, Burt
D.
Plasma interleukin-6, tumour necrosis factor alpha and blood cytokine production in type 2 diabetes
.
Life Sci
2000
;
67
:
291
–
300
.
307Donath
MY
, Shoelson
SE.
Type 2 diabetes as an inflammatory disease
.
Nat Rev Immunol
2011
;
11
:
98
–
107
.
308Dinh
W
, Futh
R
, Nickl
W
, Krahn
T
, Ellinghaus
P
, Scheffold
T
, Bansemir
L
, Bufe
A
, Barroso
MC
, Lankisch
M.
Elevated plasma levels of TNF-alpha and interleukin-6 in patients with diastolic dysfunction and glucose metabolism disorders
.
Cardiovasc Diabetol
2009
;
8
:
58
.
309Marwick
TH
, Ritchie
R
, Shaw
JE
, Kaye
D.
Implications of underlying mechanisms for the recognition and management of diabetic cardiomyopathy
.
J Am Coll Cardiol
2018
;
71
:
339
–
351
.
310Frieler
RA
, Mortensen
RM.
Immune cell and other noncardiomyocyte regulation of cardiac hypertrophy and remodeling
.
Circulation
2015
;
131
:
1019
–
1030
.
311Mann
DL.
Innate immunity and the failing heart: the cytokine hypothesis revisited
.
Circ Res
2015
;
116
:
1254
–
1268
.
312Tan
Y
, Zhang
Z
, Zheng
C
, Wintergerst
KA
, Keller
BB
, Cai
L.
Mechanisms of diabetic cardiomyopathy and potential therapeutic strategies: preclinical and clinical evidence
.
Nat Rev Cardiol
2020
;
17
:
585
–
607
.
313Ha
T
, Li
Y
, Hua
F
, Ma
J
, Gao
X
, Kelley
J
, Zhao
A
, Haddad
GE
, Williams
DL
, William Browder
I
, Kao
RL
, Li
C.
Reduced cardiac hypertrophy in toll-like receptor 4-deficient mice following pressure overload
.
Cardiovasc Res
2005
;
68
:
224
–
234
.
314Tao
A
, Song
J
, Lan
T
, Xu
X
, Kvietys
P
, Kao
R
, Martin
C
, Rui
T.
Cardiomyocyte-fibroblast interaction contributes to diabetic cardiomyopathy in mice: role of HMGB1/TLR4/IL-33 axis
.
Biochim Biophys Acta
2015
;
1852
:
2075
–
2085
.
315Lorenzo
O
, Picatoste
B
, Ares-Carrasco
S
, Ramirez
E
, Egido
J
, Tunon
J.
Potential role of nuclear factor kappaB in diabetic cardiomyopathy
.
Mediators Inflamm
2011
;
2011
:
652097
.
316Gordon
JW
, Shaw
JA
, Kirshenbaum
LA.
Multiple facets of NF-kappaB in the heart: to be or not to NF-kappaB
.
Circ Res
2011
;
108
:
1122
–
1132
.
317Frati
G
, Schirone
L
, Chimenti
I
, Yee
D
, Biondi-Zoccai
G
, Volpe
M
, Sciarretta
S.
An overview of the inflammatory signalling mechanisms in the myocardium underlying the development of diabetic cardiomyopathy
.
Cardiovasc Res
2017
;
113
:
378
–
388
.
318Varga
ZV
, Giricz
Z
, Liaudet
L
, Hasko
G
, Ferdinandy
P
, Pacher
P.
Interplay of oxidative, nitrosative/nitrative stress, inflammation, cell death and autophagy in diabetic cardiomyopathy
.
Biochim Biophys Acta
2015
;
1852
:
232
–
242
.
319Palomer
X
, Salvado
L
, Barroso
E
, Vazquez-Carrera
M.
An overview of the crosstalk between inflammatory processes and metabolic dysregulation during diabetic cardiomyopathy
.
Int J Cardiol
2013
;
168
:
3160
–
3172
.
320Condorelli
G
, Morisco
C
, Latronico
MV
, Claudio
PP
, Dent
P
, Tsichlis
P
, Condorelli
G
, Frati
G
, Drusco
A
, Croce
CM
, Napoli
C.
TNF-alpha signal transduction in rat neonatal cardiac myocytes: definition of pathways generating from the TNF-alpha receptor
.
FASEB J
2002
;
16
:
1732
–
1737
.
321Kawano
S
, Kubota
T
, Monden
Y
, Kawamura
N
, Tsutsui
H
, Takeshita
A
, Sunagawa
K.
Blockade of NF-kappaB ameliorates myocardial hypertrophy in response to chronic infusion of angiotensin II
.
Cardiovasc Res
2005
;
67
:
689
–
698
.
322Honsho
S
, Nishikawa
S
, Amano
K
, Zen
K
, Adachi
Y
, Kishita
E
, Matsui
A
, Katsume
A
, Yamaguchi
S
, Nishikawa
K
, Isoda
K
, Riches
DW
, Matoba
S
, Okigaki
M
, Matsubara
H.
Pressure-mediated hypertrophy and mechanical stretch induces IL-1 release and subsequent IGF-1 generation to maintain compensative hypertrophy by affecting Akt and JNK pathways
.
Circ Res
2009
;
105
:
1149
–
1158
.
323Zhao
L
, Cheng
G
, Jin
R
, Afzal
MR
, Samanta
A
, Xuan
YT
, Girgis
M
, Elias
HK
, Zhu
Y
, Davani
A
, Yang
Y
, Chen
X
, Ye
S
, Wang
OL
, Chen
L
, Hauptman
J
, Vincent
RJ
, Dawn
B.
Deletion of interleukin-6 attenuates pressure overload-induced left ventricular hypertrophy and dysfunction
.
Circ Res
2016
;
118
:
1918
–
1929
.
324Matsumoto-Ida
M
, Takimoto
Y
, Aoyama
T
, Akao
M
, Takeda
T
, Kita
T.
Activation of TGF-beta1-TAK1-p38 MAPK pathway in spared cardiomyocytes is involved in left ventricular remodeling after myocardial infarction in rats
.
Am J Physiol Heart Circ Physiol
2006
;
290
:
H709
–
H715
.
325Lim
JY
, Park
SJ
, Hwang
HY
, Park
EJ
, Nam
JH
, Kim
J
, Park
SI.
TGF-beta1 induces cardiac hypertrophic responses via PKC-dependent ATF-2 activation
.
J Mol Cell Cardiol
2005
;
39
:
627
–
636
.
326Haudek
SB
, Taffet
GE
, Schneider
MD
, Mann
DL.
TNF provokes cardiomyocyte apoptosis and cardiac remodeling through activation of multiple cell death pathways
.
J Clin Invest
2007
;
117
:
2692
–
2701
.
327Kawamura
N
, Kubota
T
, Kawano
S
, Monden
Y
, Feldman
AM
, Tsutsui
H
, Takeshita
A
, Sunagawa
K.
Blockade of NF-kappaB improves cardiac function and survival without affecting inflammation in TNF-alpha-induced cardiomyopathy
.
Cardiovasc Res
2005
;
66
:
520
–
529
.
328Liu
Z
, Zhao
N
, Zhu
H
, Zhu
S
, Pan
S
, Xu
J
, Zhang
X
, Zhang
Y
, Wang
J.
Circulating interleukin-1beta promotes endoplasmic reticulum stress-induced myocytes apoptosis in diabetic cardiomyopathy via interleukin-1 receptor-associated kinase-2
.
Cardiovasc Diabetol
2015
;
14
:
125
.
329Mezzaroma
E
, Toldo
S
, Farkas
D
, Seropian
IM
, Van Tassell
BW
, Salloum
FN
, Kannan
HR
, Menna
AC
, Voelkel
NF
, Abbate
A.
The inflammasome promotes adverse cardiac remodeling following acute myocardial infarction in the mouse
.
Proc Natl Acad Sci USA
2011
;
108
:
19725
–
19730
.
330Xie
Y
, Huang
Y
, Ling
X
, Qin
H
, Wang
M
, Luo
B.
Chemerin/CMKLR1 axis promotes inflammation and pyroptosis by activating NLRP3 inflammasome in diabetic cardiomyopathy rat
.
Front Physiol
2020
;
11
:
1
–13.
331Zhang
Y
, Wang
JH
, Zhang
YY
, Wang
YZ
, Wang
J
, Zhao
Y
, Jin
XX
, Xue
GL
, Li
PH
, Sun
YL
, Huang
QH
, Song
XT
, Zhang
ZR
, Gao
X
, Yang
BF
, Du
ZM
, Pan
ZW.
Deletion of interleukin-6 alleviated interstitial fibrosis in streptozotocin-induced diabetic cardiomyopathy of mice through affecting TGFbeta1 and miR-29 pathways
.
Sci Rep
2016
;
6
:
23010
.
332Venkatachalam
K
, Venkatesan
B
, Valente
AJ
, Melby
PC
, Nandish
S
, Reusch
JE
, Clark
RA
, Chandrasekar
B.
WISP1, a pro-mitogenic, pro-survival factor, mediates tumor necrosis factor-alpha (TNF-alpha)-stimulated cardiac fibroblast proliferation but inhibits TNF-alpha-induced cardiomyocyte death
.
J Biol Chem
2009
;
284
:
14414
–
14427
.
333Ko
HJ
, Zhang
Z
, Jung
DY
, Jun
JY
, Ma
Z
, Jones
KE
, Chan
SY
, Kim
JK.
Nutrient stress activates inflammation and reduces glucose metabolism by suppressing AMP-activated protein kinase in the heart
.
Diabetes
2009
;
58
:
2536
–
2546
.
334Palomer
X
, Alvarez-Guardia
D
, Rodriguez-Calvo
R
, Coll
T
, Laguna
JC
, Davidson
MM
, Chan
TO
, Feldman
AM
, Vazquez-Carrera
M.
TNF-alpha reduces PGC-1alpha expression through NF-kappaB and p38 MAPK leading to increased glucose oxidation in a human cardiac cell model
.
Cardiovasc Res
2009
;
81
:
703
–
712
.
335Alvarez-Guardia
D
, Palomer
X
, Coll
T
, Davidson
MM
, Chan
TO
, Feldman
AM
, Laguna
JC
, Vazquez-Carrera
M.
The p65 subunit of NF-kappaB binds to PGC-1alpha, linking inflammation and metabolic disturbances in cardiac cells
.
Cardiovasc Res
2010
;
87
:
449
–
458
.
336Bajpai
A
, Tilley
DG.
The role of leukocytes in diabetic cardiomyopathy
.
Front Physiol
2018
;
9
:
1
–12.
337Thaik
CM
, Calderone
A
, Takahashi
N
, Colucci
WS.
Interleukin-1 beta modulates the growth and phenotype of neonatal rat cardiac myocytes
.
J Clin Invest
1995
;
96
:
1093
–
1099
.
338Finkel
MS
, Oddis
CV
, Jacob
TD
, Watkins
SC
, Hattler
BG
, Simmons
RL.
Negative inotropic effects of cytokines on the heart mediated by nitric oxide
.
Science
1992
;
257
:
387
–
389
.
339Noh
H
, Yu
MR
, Kim
HJ
, Lee
JH
, Park
BW
, Wu
IH
, Matsumoto
M
, King
GL.
Beta 2-adrenergic receptor agonists are novel regulators of macrophage activation in diabetic renal and cardiovascular complications
.
Kidney Int
2017
;
92
:
101
–
113
.
340Falcao-Pires
I
, Leite-Moreira
AF.
Diabetic cardiomyopathy: understanding the molecular and cellular basis to progress in diagnosis and treatment
.
Heart Fail Rev
2012
;
17
:
325
–
344
.
341Zhou
X
, Ma
L
, Habibi
J
, Whaley-Connell
A
, Hayden
MR
, Tilmon
RD
, Brown
AN
, Kim
JA
, Demarco
VG
, Sowers
JR.
Nebivolol improves diastolic dysfunction and myocardial remodeling through reductions in oxidative stress in the Zucker obese rat
.
Hypertension
2010
;
55
:
880
–
888
.
342Hayden
MR
, Habibi
J
, Joginpally
T
, Karuparthi
PR
, Sowers
JR.
Ultrastructure study of transgenic Ren2 rat aorta—part 1: endothelium and Intima
.
Cardiorenal Med
2012
;
2
:
66
–
82
.
343Nozynski
J
, Zakliczynski
M
, Konecka-Mrowka
D
, Przybylski
R
, Zembala
M
, Zielinska
T
, Mrowka
A
, Lange
D
, Zembala-Nozynska
E
, Nikiel
B
, Mlynarczyk-Liszka
J.
Advanced glycation end-products in myocardium-supported vessels: effects of heart failure and diabetes mellitus
.
J Heart Lung Transplant
2011
;
30
:
558
–
564
.
344Bodiga
VL
, Eda
SR
, Bodiga
S.
Advanced glycation end products: role in pathology of diabetic cardiomyopathy
.
Heart Fail Rev
2014
;
19
:
49
–
63
.
345Kumari
K
, Sahib
MK.
Susceptibility of different rat tissues to non-enzymatic protein glycosylation in experimental diabetes
.
Indian J Exp Biol
1993
;
31
:
194
–
195
.
346Nożyński
J
, Zakliczyński
M
, Konecka-Mrowka
D
, Zielinska
T
, Zakliczynska
H
, Nikiel
B
, Mlynarczyk-Liszka
J
, Mrowka
A
, Zembala-Nozynska
E
, Pijet
M
, Rdzanowska
K
, Lange
D
, Przybylski
R
, Zembala
M.
Advanced glycation end product accumulation in the cardiomyocytes of heart failure patients with and without diabetes
.
Ann Transplant
2012
;
17
:
53
–
61
.
347Schalkwijk
CG
, Baidoshvili
A
, Stehouwer
CD
, van Hinsbergh
VW
, Niessen
HW.
Increased accumulation of the glycoxidation product Nepsilon-(carboxymethyl)lysine in hearts of diabetic patients: generation and characterisation of a monoclonal anti-CML antibody
.
Biochim Biophys Acta
2004
;
1636
:
82
–
89
.
348Candido
R
, Forbes
JM
, Thomas
MC
, Thallas
V
, Dean
RG
, Burns
WC
, Tikellis
C
, Ritchie
RH
, Twigg
SM
, Cooper
ME
, Burrell
LM.
A breaker of advanced glycation end products attenuates diabetes-induced myocardial structural changes
.
Circ Res
2003
;
92
:
785
–
792
.
349Liu
J
, Masurekar
MR
, Vatner
DE
, Jyothirmayi
GN
, Regan
TJ
, Vatner
SF
, Meggs
LG
, Malhotra
A.
Glycation end-product cross-link breaker reduces collagen and improves cardiac function in aging diabetic heart
.
Am J Physiol Heart Circ Physiol
2003
;
285
:
H2587
–
2591
.
350Yilmaz
S
, Canpolat
U
, Aydogdu
S
, Abboud
HE.
Diabetic cardiomyopathy; summary of 41 years
.
Korean Circ J
2015
;
45
:
266
–
272
.
351Ko
SY
, Chang
SS
, Lin
IH
, Chen
HI.
Suppression of antioxidant Nrf-2 and downstream pathway in H9c2 cells by advanced glycation end products (AGEs) via ERK phosphorylation
.
Biochimie
2015
;
118
:
8
–
14
.
352Hou
J
, Zheng
D
, Fung
G
, Deng
H
, Chen
L
, Liang
J
, Jiang
Y
, Hu
Y.
Mangiferin suppressed advanced glycation end products (AGEs) through NF-κB deactivation and displayed anti-inflammatory effects in streptozotocin and high fat diet-diabetic cardiomyopathy rats
.
Can J Physiol Pharmacol
2016
;
94
:
332
–
340
.
353Yuan
Q
, Zhou
QY
, Liu
D
, Yu
L
, Zhan
L
, Li
XJ
, Peng
HY
, Zhang
XL
, Yuan
XC.
Advanced glycation end-products impair Na(+)/K(+)-ATPase activity in diabetic cardiomyopathy: role of the adenosine monophosphate-activated protein kinase/sirtuin 1 pathway
.
Clin Exp Pharmacol Physiol
2014
;
41
:
127
–
133
.
354Aragno
M
, Mastrocola
R
, Medana
C
, Catalano
MG
, Vercellinatto
I
, Danni
O
, Boccuzzi
G.
Oxidative stress-dependent impairment of cardiac-specific transcription factors in experimental diabetes
.
Endocrinology
2006
;
147
:
5967
–
5974
.
355Ma
H
, Li
SY
, Xu
P
, Babcock
SA
, Dolence
EK
, Brownlee
M
, Li
J
, Ren
J.
Advanced glycation endproduct (AGE) accumulation and AGE receptor (RAGE) up-regulation contribute to the onset of diabetic cardiomyopathy
.
J Cell Mol Med
2009
;
13
:
1751
–
1764
.
356Nielsen
JM
, Kristiansen
SB
, Norregaard
R
, Andersen
CL
, Denner
L
, Nielsen
TT
, Flyvbjerg
A
, Botker
HE.
Blockage of receptor for advanced glycation end products prevents development of cardiac dysfunction in db/db type 2 diabetic mice
.
Eur J Heart Fail
2009
;
11
:
638
–
647
.
357Torres
CR
, Hart
GW.
Topography and polypeptide distribution of terminal N-acetylglucosamine residues on the surfaces of intact lymphocytes. Evidence for O-linked GlcNAc
.
J Biol Chem
1984
;
259
:
3308
–
3317
.
358Marshall
S
, Bacote
V
, Traxinger
RR.
Discovery of a metabolic pathway mediating glucose-induced desensitization of the glucose transport system. Role of hexosamine biosynthesis in the induction of insulin resistance
.
J Biol Chem
1991
;
266
:
4706
–
4712
.
359Gibb
AA
, Lorkiewicz
PK
, Zheng
Y-T
, Zhang
X
, Bhatnagar
A
, Jones
SP
, Hill
BG.
Integration of flux measurements to resolve changes in anabolic and catabolic metabolism in cardiac myocytes
.
Biochem J
2017
;
474
:
2785
–
2801
.
360Olson
AK
, Bouchard
B
, Zhu
WZ
, Chatham
JC.
Des Rosiers C. First characterization of glucose flux through the hexosamine biosynthesis pathway (HBP) in ex vivo mouse heart
.
J Biol Chem
2020
;
295
:
2018
–
2033
.
361Hart
GW
, Slawson
C
, Ramirez-Correa
G
, Lagerlof
O.
Cross talk between O-GlcNAcylation and phosphorylation: roles in signaling, transcription, and chronic disease
.
Annu Rev Biochem
2011
;
80
:
825
–
858
.
362Snow
CM
, Senior
A
, Gerace
L.
Monoclonal antibodies identify a group of nuclear pore complex glycoproteins
.
J Cell Biol
1987
;
104
:
1143
–
1156
.
363Chatham
JC
, Young
ME
, Zhang
JH.
Reprint of: role of O-linked N-acetylglucosamine (O-GlcNAc) modification of proteins in diabetic cardiovascular complications
.
Curr Opin Pharm
2020
;
54
:
209
–
220
.
364Prakoso
D
, Kiriazis
H
, Tate
M
, Qian
H
, Deo
M
, Parry
L
, Gregorevic
P
, Du
X
, Chatham
J
, De Blasio
M.
5213 manipulation of cardiac O-GlcNAc modification alters cardiac function and remodelling in the setting of diabetic cardiomyopathy
.
Eur Heart J
2018
;
39
:ehy566.5213.
365Erickson
JR
, Pereira
L
, Wang
L
, Han
G
, Ferguson
A
, Dao
K
, Copeland
RJ
, Despa
F
, Hart
GW
, Ripplinger
CM
, Bers
DM.
Diabetic hyperglycaemia activates CaMKII and arrhythmias by O-linked glycosylation
.
Nature
2013
;
502
:
372
–
376
.
366Lehman
DM
, Fu
D-J
, Freeman
AB
, Hunt
KJ
, Leach
RJ
, Johnson-Pais
T
, Hamlington
J
, Dyer
TD
, Arya
R
, Abboud
H.
A single nucleotide polymorphism in mgea5 encoding o-glcnac–selective n-acetyl-β-d glucosaminidase is associated with type 2 diabetes in mexican americans
.
Diabetes
2005
;
54
:
1214
–
1221
.
367Duggirala
R
, Blangero
J
, Almasy
L
, Dyer
TD
, Williams
KL
, Leach
RJ
, O'Connell
P
, Stern
MP.
Linkage of type 2 diabetes mellitus and of age at onset to a genetic location on chromosome 10q in Mexican Americans
.
Am J Hum Genet
1999
;
64
:
1127
–
1140
.
368Han
I
, Kudlow
JE.
Reduced O glycosylation of Sp1 is associated with increased proteasome susceptibility
.
Mol Cell Biol
1997
;
17
:
2550
–
2558
.
369Du
XL
, Edelstein
D
, Dimmeler
S
, Ju
QD
, Sui
C
, Brownlee
M.
Hyperglycemia inhibits endothelial nitric oxide synthase activity by posttranslational modification at the Akt site
.
J Clin Invest
2001
;
108
:
1341
–
1348
.
370Akimoto
Y
, Kreppel
LK
, Hirano
H
, Hart
GW.
Hyperglycemia and the O-GlcNAc transferase in rat aortic smooth muscle cells: elevated expression and altered patterns of O-GlcNAcylation
.
Arch Biochem Biophys
2001
;
389
:
166
–
175
.
371Ma
J
, Banerjee
P
, Whelan
SA
, Liu
T
, Wei
AC
, Ramirez-Correa
G
, McComb
ME
, Costello
CE
, O'Rourke
B
, Murphy
A
, Hart
GW.
Comparative proteomics reveals dysregulated mitochondrial O-GlcNAcylation in diabetic hearts
.
J Proteome Res
2016
;
15
:
2254
–
2264
.
372Ramirez-Correa
GA
, Ma
J
, Slawson
C
, Zeidan
Q
, Lugo-Fagundo
NS
, Xu
M
, Shen
X
, Gao
WD
, Caceres
V
, Chakir
K
, DeVine
L
, Cole
RN
, Marchionni
L
, Paolocci
N
, Hart
GW
, Murphy
AM.
Removal of abnormal myofilament O-GlcNAcylation restores Ca2+ sensitivity in diabetic cardiac muscle
.
Diabetes
2015
;
64
:
3573
–
3587
.
373Banerjee
PS
, Ma
J
, Hart
GW.
Diabetes-associated dysregulation of O-GlcNAcylation in rat cardiac mitochondria
.
Proc Natl Acad Sci USA
2015
;
112
:
6050
–
6055
.
374Cao
H
, Hu
Y
, Zhu
X
, Yao
N
, Gu
J
, Wang
Y
, Zhu
W.
O-GlcNAc transferase affects the signal transduction of beta1 adrenoceptor in adult rat cardiomyocytes by increasing the O-GlcNAcylation of beta1 adrenoceptor
.
Biochem Biophys Res Commun
2020
;
528
:
71
–
77
.
375Kronlage
M
, Dewenter
M
, Grosso
J
, Fleming
T
, Oehl
U
, Lehmann
LH
, Falcão-Pires
I
, Leite-Moreira
AF
, Volk
N
, Gröne
H-J
, Müller
OJ
, Sickmann
A
, Katus
HA
, Backs
J.
O-GlcNAcylation of histone deacetylase 4 protects the diabetic heart from failure
.
Circulation
2019
;
140
:
580
–
594
.
376Huang
L
, Yuan
P
, Yu
P
, Kong
Q
, Xu
Z
, Yan
X
, Shen
Y
, Yang
J
, Wan
R
, Hong
K
, Tang
Y
, Hu
J.
O-GlcNAc-modified SNAP29 inhibits autophagy-mediated degradation via the disturbed SNAP29-STX17-VAMP8 complex and exacerbates myocardial injury in type I diabetic rats
.
Int J Mol Med
2018
;
42
:
3278
–
3290
.
377Hegyi
B
, Borst
JM
, Bailey
LRJ
, Shen
EY
, Lucena
AJ
, Navedo
MF
, Bossuyt
J
, Bers
DM.
Hyperglycemia regulates cardiac K(+) channels via O-GlcNAc-CaMKII and NOX2-ROS-PKC pathways
.
Basic Res Cardiol
2020
;
115
:
71
.
378Yu
P
, Hu
L
, Xie
J
, Chen
S
, Huang
L
, Xu
Z
, Liu
X
, Zhou
Q
, Yuan
P
, Yan
X
, Jin
J
, Shen
Y
, Zhu
W
, Fu
L
, Chen
Q
, Yu
J
, Hu
J
, Cao
Q
, Wan
R
, Hong
K.
O-GlcNAcylation of cardiac Nav1.5 contributes to the development of arrhythmias in diabetic hearts
.
Int J Cardiol
2018
;
260
:
74
–
81
.
379Masaki
N
, Feng
B
, Breton-Romero
R
, Inagaki
E
, Weisbrod
RM
, Fetterman
JL
, Hamburg
NM.
O-GlcNAcylation mediates glucose-induced alterations in endothelial cell phenotype in human diabetes mellitus
.
J Am Heart Assoc
2020
;
9
:
e014046
.
380Frustaci
A
, Kajstura
J
, Chimenti
C
, Jakoniuk
I
, Leri
A
, Maseri
A
, Nadal-Ginard
B
, Anversa
P.
Myocardial cell death in human diabetes
.
Circ Res
2000
;
87
:
1123
–
1132
.
381Riehle
C
, Bauersachs
J.
Of mice and men: models and mechanisms of diabetic cardiomyopathy
.
Basic Res Cardiol
2019
;
114
:
2
.
382Selvin
E
, Lazo
M
, Chen
Y
, Shen
L
, Rubin
J
, McEvoy
JW
, Hoogeveen
RC
, Sharrett
AR
, Ballantyne
CM
, Coresh
J.
Diabetes mellitus, prediabetes, and incidence of subclinical myocardial damage
.
Circulation
2014
;
130
:
1374
–
1382
.
383Hou
N
, Mai
Y
, Qiu
X
, Yuan
W
, Li
Y
, Luo
C
, Liu
Y
, Zhang
G
, Zhao
G
, Luo
JD.
Carvacrol attenuates diabetic cardiomyopathy by modulating the PI3K/AKT/GLUT4 pathway in diabetic mice
.
Front Pharmacol
2019
;
10
:
1
–15.
384Olivetti
G
, Abbi
R
, Quaini
F
, Kajstura
J
, Cheng
W
, Nitahara
JA
, Quaini
E
, Di Loreto
C
, Beltrami
CA
, Krajewski
S
, Reed
JC
, Anversa
P.
Apoptosis in the failing human heart
.
N Engl J Med
1997
;
336
:
1131
–
1141
.
385Wencker
D
, Chandra
M
, Nguyen
K
, Miao
W
, Garantziotis
S
, Factor
SM
, Shirani
J
, Armstrong
RC
, Kitsis
RN.
A mechanistic role for cardiac myocyte apoptosis in heart failure
.
J Clin Invest
2003
;
111
:
1497
–
1504
.
386von Harsdorf
R
, Li
PF
, Dietz
R.
Signaling pathways in reactive oxygen species-induced cardiomyocyte apoptosis
.
Circulation
1999
;
99
:
2934
–
2941
.
387Xu
J
, Wang
G
, Wang
Y
, Liu
Q
, Xu
W
, Tan
Y
, Cai
L.
Diabetes- and angiotensin II-induced cardiac endoplasmic reticulum stress and cell death: metallothionein protection
.
J Cell Mol Med
2009
;
13
:
1499
–
1512
.
388Zou
H
, Henzel
WJ
, Liu
X
, Lutschg
A
, Wang
X.
Apaf-1, a human protein homologous to C. elegans CED-4, participates in cytochrome c-dependent activation of caspase-3
.
Cell
1997
;
90
:
405
–
413
.
389Li
P
, Nijhawan
D
, Budihardjo
I
, Srinivasula
SM
, Ahmad
M
, Alnemri
ES
, Wang
X.
Cytochrome c and dATP-dependent formation of Apaf-1/caspase-9 complex initiates an apoptotic protease cascade
.
Cell
1997
;
91
:
479
–
489
.
390Stennicke
HR
, Deveraux
QL
, Humke
EW
, Reed
JC
, Dixit
VM
, Salvesen
GS.
Caspase-9 can be activated without proteolytic processing
.
J Biol Chem
1999
;
274
:
8359
–
8362
.
391Renatus
M
, Stennicke
HR
, Scott
FL
, Liddington
RC
, Salvesen
GS.
Dimer formation drives the activation of the cell death protease caspase 9
.
Proc Natl Acad Sci USA
2001
;
98
:
14250
–
14255
.
392Toshiyuki
M
, Reed
JC.
Tumor suppressor p53 is a direct transcriptional activator of the human bax gene
.
Cell
1995
;
80
:
293
–
299
.
393Oltval
ZN
, Milliman
CL
, Korsmeyer
SJ.
Bcl-2 heterodimerizes in vivo with a conserved homolog, Bax, that accelerates programed cell death
.
Cell
1993
;
74
:
609
–
619
.
394Siegel
RM
, Frederiksen
JK
, Zacharias
DA
, Chan
FK
, Johnson
M
, Lynch
D
, Tsien
RY
, Lenardo
MJ.
Fas preassociation required for apoptosis signaling and dominant inhibition by pathogenic mutations
.
Science
2000
;
288
:
2354
–
2357
.
395Kischkel
FC
, Hellbardt
S
, Behrmann
I
, Germer
M
, Pawlita
M
, Krammer
PH
, Peter
ME.
Cytotoxicity-dependent APO-1 (Fas/CD95)-associated proteins form a death-inducing signaling complex (DISC) with the receptor
.
EMBO J
1995
;
14
:
5579
–
5588
.
396Boldin
MP
, Goncharov
TM
, Goltsev
YV
, Wallach
D.
Involvement of MACH, a novel MORT1/FADD-interacting protease, in Fas/APO-1- and TNF receptor-induced cell death
.
Cell
1996
;
85
:
803
–
815
.
397Boatright
KM
, Renatus
M
, Scott
FL
, Sperandio
S
, Shin
H
, Pedersen
IM
, Ricci
JE
, Edris
WA
, Sutherlin
DP
, Green
DR
, Salvesen
GS.
A unified model for apical caspase activation
.
Mol Cell
2003
;
11
:
529
–
541
.
398Luo
X
, Budihardjo
I
, Zou
H
, Slaughter
C
, Wang
X.
Bid, a Bcl2 interacting protein, mediates cytochrome c release from mitochondria in response to activation of cell surface death receptors
.
Cell
1998
;
94
:
481
–
490
.
399Takemura
G
, Kanoh
M
, Minatoguchi
S
, Fujiwara
H.
Cardiomyocyte apoptosis in the failing heart--a critical review from definition and classification of cell death
.
Int J Cardiol
2013
;
167
:
2373
–
2386
.
400Kostin
S
, Pool
L
, ElsäSser
A
, Hein
S
, Drexler
HCA
, Arnon
E
, Hayakawa
Y
, Zimmermann
R
, Bauer
E
, KlöVekorn
W-P
, Schaper
J.
Myocytes die by multiple mechanisms in failing human hearts
.
Circ Res
2003
;
92
:
715
–
724
.
401Hein
S
, Arnon
E
, Kostin
S
, SchöNburg
M
, ElsäSser
A
, Polyakova
V
, Bauer
EP
, KlöVekorn
W-P
, Schaper
J
. Progression from compensated hypertrophy to failure in the pressure-overloaded human heart: structural deterioration and compensatory mechanisms
.
Circulation
2003
;
107
:
984
–
991
.
402Nakagami
H
, Morishita
R
, Yamamoto
K
, Yoshimura
SI
, Taniyama
Y
, Aoki
M
, Matsubara
H
, Kim
S
, Kaneda
Y
, Ogihara
T.
Phosphorylation of p38 mitogen-activated protein kinase downstream of bax-caspase-3 pathway leads to cell death induced by high D-glucose in human endothelial cells
.
Diabetes
2001
;
50
:
1472
–
1481
.
403Cai
L
, Li
W
, Wang
G
, Guo
L
, Jiang
Y
, Kang
YJ.
Hyperglycemia-induced apoptosis in mouse myocardium: mitochondrial cytochrome C-mediated caspase-3 activation pathway
.
Diabetes
2002
;
51
:
1938
–
1948
.
404Monkemann
H
, De Vriese
AS
, Blom
HJ
, Kluijtmans
LA
, Heil
SG
, Schild
HH
, Golubnitschaja
O.
Early molecular events in the development of the diabetic cardiomyopathy
.
Amino Acids
2002
;
23
:
331
–
336
.
405Kajstura
J
, Fiordaliso
F
, Andreoli
AM
, Li
B
, Chimenti
S
, Medow
MS
, Limana
F
, Nadal-Ginard
B
, Leri
A
, Anversa
P.
IGF-1 overexpression inhibits the development of diabetic cardiomyopathy and angiotensin II-mediated oxidative stress
.
Diabetes
2001
;
50
:
1414
–
1424
.
406Sparagna
GC
, Hickson-Bick
DL
, Buja
LM
, McMillin
JB.
A metabolic role for mitochondria in palmitate-induced cardiac myocyte apoptosis
.
Am J Physiol Heart Circ Physiol
2000
;
279
:
H2124
–
2132
.
407Ohkuwa
T
, Sato
Y
, Naoi
M.
Hydroxyl radical formation in diabetic rats induced by streptozotocin
.
Life Sci
1995
;
56
:
1789
–
1798
.
408Shizukuda
Y
, Reyland
ME
, Buttrick
PM.
Protein kinase C-delta modulates apoptosis induced by hyperglycemia in adult ventricular myocytes
.
Am J Physiol Heart Circ Physiol
2002
;
282
:
H1625
–
1634
.
409Bryant
D
, Becker
L
, Richardson
J
, Shelton
J
, Franco
F
, Peshock
R
, Thompson
M
, Giroir
B.
Cardiac failure in transgenic mice with myocardial expression of tumor necrosis factor-alpha
.
Circulation
1998
;
97
:
1375
–
1381
.
410Sciarretta
S
, Maejima
Y
, Zablocki
D
, Sadoshima
J.
The role of autophagy in the heart
.
Annu Rev Physiol
2018
;
80
:
1
–
26
.
411Riehle
C
, Wende
AR
, Sena
S
, Pires
KM
, Pereira
RO
, Zhu
Y
, Bugger
H
, Frank
D
, Bevins
J
, Chen
D
, Perry
CN
, Dong
XC
, Valdez
S
, Rech
M
, Sheng
X
, Weimer
BC
, Gottlieb
RA
, White
MF
, Abel
ED.
Insulin receptor substrate signaling suppresses neonatal autophagy in the heart
.
J Clin Invest
2013
;
123
:
5319
–
5333
.
412Pires
KM
, Torres
NS
, Buffolo
M
, Gunville
R
, Schaaf
C
, Davis
K
, Selzman
CH
, Gottlieb
RA
, Boudina
S.
Suppression of cardiac autophagy by hyperinsulinemia in insulin receptor-deficient hearts is mediated by insulin-like growth factor receptor signaling
.
Antioxid Redox Signal
2019
;
31
:
444
–
457
.
413Xie
ZL
, Lau
K
, Eby
B
, Lozano
P
, He
CY
, Pennington
B
, Li
HL
, Rathi
S
, Dong
YZ
, Tian
R
, Kem
D
, Zou
MH.
Improvement of cardiac functions by chronic metformin treatment is associated with enhanced cardiac autophagy in diabetic OVE26 Mice
.
Diabetes
2011
;
60
:
1770
–
1778
.
414Pires
KM
, Buffolo
M
, Schaaf
C
, Symons
JD
, Cox
J
, Abel
ED
, Selzman
CH
, Boudina
S.
Activation of IGF-1 receptors and Akt signaling by systemic hyperinsulinemia contributes to cardiac hypertrophy but does not regulate cardiac autophagy in obese diabetic mice
.
J Mol Cell Cardiol
2017
;
113
:
39
–
50
.
415Zhao
Y
, Zhang
L
, Qiao
Y
, Zhou
X
, Wu
G
, Wang
L
, Peng
Y
, Dong
X
, Huang
H
, Si
L
, Zhang
X
, Zhang
L
, Li
J
, Wang
W
, Zhou
L
, Gao
X.
Heme oxygenase-1 prevents cardiac dysfunction in streptozotocin-diabetic mice by reducing inflammation, oxidative stress, apoptosis and enhancing autophagy
.
PLoS One
2013
;
8
:
e75927
.
416Xie
Z
, He
C
, Zou
MH.
AMP-activated protein kinase modulates cardiac autophagy in diabetic cardiomyopathy
.
Autophagy
2011
;
7
:
1254
–
1255
.
417Das
A
, Durrant
D
, Koka
S
, Salloum
FN
, Xi
L
, Kukreja
RC.
Mammalian target of rapamycin (mTOR) inhibition with rapamycin improves cardiac function in type 2 diabetic mice: potential role of attenuated oxidative stress and altered contractile protein expression
.
J Biol Chem
2014
;
289
:
4145
–
4160
.
418Morselli
E
, Maiuri
MC
, Markaki
M
, Megalou
E
, Pasparaki
A
, Palikaras
K
, Criollo
A
, Galluzzi
L
, Malik
SA
, Vitale
I
, Michaud
M
, Madeo
F
, Tavernarakis
N
, Kroemer
G.
Caloric restriction and resveratrol promote longevity through the Sirtuin-1-dependent induction of autophagy
.
Cell Death Dis
2010
;
1
:
e10
.
419Zhang
M
, Zhang
L
, Hu
J
, Lin
J
, Wang
T
, Duan
Y
, Man
W
, Feng
J
, Sun
L
, Jia
H
, Li
C
, Zhang
R
, Wang
H
, Sun
D.
MST1 coordinately regulates autophagy and apoptosis in diabetic cardiomyopathy in mice
.
Diabetologia
2016
;
59
:
2435
–
2447
.
420Nakayama
H
, Chen
X
, Baines
CP
, Klevitsky
R
, Zhang
X
, Zhang
H
, Jaleel
N
, Chua
BH
, Hewett
TE
, Robbins
J
, Houser
SR
, Molkentin
JD.
Ca2+- and mitochondrial-dependent cardiomyocyte necrosis as a primary mediator of heart failure
.
J Clin Invest
2007
;
117
:
2431
–
2444
.
421Lu
LQ
, Tian
J
, Luo
XJ
, Peng
J.
Targeting the pathways of regulated necrosis: a potential strategy for alleviation of cardio-cerebrovascular injury
.
Cell Mol Life Sci
2021
;
78
:
63
–
78
.
422Wang
X
, Pan
J
, Liu
H
, Zhang
M
, Liu
D
, Lu
L
, Tian
J
, Liu
M
, Jin
T
, An
F.
AIM2 gene silencing attenuates diabetic cardiomyopathy in type 2 diabetic rat model
.
Life Sci
2019
;
221
:
249
–
258
.
423Yang
F
, Qin
Y
, Lv
J
, Wang
Y
, Che
H
, Chen
X
, Jiang
Y
, Li
A
, Sun
X
, Yue
E
, Ren
L
, Li
Y
, Bai
Y
, Wang
L.
Silencing long non-coding RNA Kcnq1ot1 alleviates pyroptosis and fibrosis in diabetic cardiomyopathy
.
Cell Death Dis
2018
;
9
:
1000
.
424Yang
F
, Qin
Y
, Wang
Y
, Meng
S
, Xian
H
, Che
H
, Lv
J
, Li
Y
, Yu
Y
, Bai
Y
, Wang
L.
Metformin inhibits the NLRP3 inflammasome via AMPK/mTOR-dependent effects in diabetic cardiomyopathy
.
Int J Biol Sci
2019
;
15
:
1010
–
1019
.
425Xie
Y
, Hou
W
, Song
X
, Yu
Y
, Huang
J
, Sun
X
, Kang
R
, Tang
D.
Ferroptosis: process and function
.
Cell Death Differ
2016
;
23
:
369
–
379
.
426Kar
S
, Shahshahan
H
, Mishra
PK.
Hydrogen sulfide protects the heart against ferroptotic cell death in diabetic cardiomyopathy
.
Circ Res
2020
;
127
:
A501
.
427Lagadic-Gossmann
D
, Buckler
KJ
, Le Prigent
K
, Feuvray
D.
Altered Ca2+ handling in ventricular myocytes isolated from diabetic rats
.
Am J Physiol
1996
;
270
:
H1529
–
1537
.
428Belke
DD
, Swanson
EA
, Dillmann
WH.
Decreased sarcoplasmic reticulum activity and contractility in diabetic db/db mouse heart
.
Diabetes
2004
;
53
:
3201
–
3208
.
429Ren
J
, Davidoff
AJ.
Diabetes rapidly induces contractile dysfunctions in isolated ventricular myocytes
.
Am J Physiol
1997
;
272
:
H148
–
H158
.
430Davidoff
AJ
, Davidson
MB
, Carmody
MW
, Davis
ME
, Ren
J.
Diabetic cardiomyocyte dysfunction and myocyte insulin resistance: role of glucose-induced PKC activity
.
Mol Cell Biochem
2004
;
262
:
155
–
163
.
431Zhou
H
, Yue
Y
, Wang
J
, Ma
Q
, Chen
Y.
Melatonin therapy for diabetic cardiomyopathy: a mechanism involving Syk-mitochondrial complex I-SERCA pathway
.
Cell Signal
2018
;
47
:
88
–
100
.
432Miki
T
, Yuda
S
, Kouzu
H
, Miura
T.
Diabetic cardiomyopathy: pathophysiology and clinical features
.
Heart Fail Rev
2013
;
18
:
149
–
166
.
433Papadaki
M
, Holewinski
RJ
, Previs
SB
, Martin
TG
, Stachowski
MJ
, Li
A
, Blair
CA
, Moravec
CS
, Van Eyk
JE
, Campbell
KS
, Warshaw
DM
, Kirk
JA.
Diabetes with heart failure increases methylglyoxal modifications in the sarcomere, which inhibit function
.
JCI Insight
2018
;
3
:
1
–17.
434Hu
Y
, Belke
D
, Suarez
J
, Swanson
E
, Clark
R
, Hoshijima
M
, Dillmann
WH.
Adenovirus-mediated overexpression of O-GlcNAcase improves contractile function in the diabetic heart
.
Circ Res
2005
;
96
:
1006
–
1013
.
435Shao
CH
, Capek
HL
, Patel
KP
, Wang
M
, Tang
K
, DeSouza
C
, Nagai
R
, Mayhan
W
, Periasamy
M
, Bidasee
KR.
Carbonylation contributes to SERCA2a activity loss and diastolic dysfunction in a rat model of type 1 diabetes
.
Diabetes
2011
;
60
:
947
–
959
.
436Kranstuber
AL
, Del
RC
, Biesiadecki
BJ
, Hamlin
RL
, Ottobre
J
, Gyorke
S
, Lacombe
VA.
Advanced glycation end product cross-link breaker attenuates diabetes-induced cardiac dysfunction by improving sarcoplasmic reticulum calcium handling
.
Front Physiol
2012
;
3
:
292
.
437Bidasee
KR
, Zhang
Y
, Shao
CH
, Wang
M
, Patel
KP
, Dincer
UD
, Besch
HR
Jr.
Diabetes increases formation of advanced glycation end products on Sarco(endo)plasmic reticulum Ca2+-ATPase
.
Diabetes
2004
;
53
:
463
–
473
.
438Pierce
GN
, Dhalla
NS.
Cardiac myofibrillar ATPase activity in diabetic rats
.
J Mol Cell Cardiol
1981
;
13
:
1063
–
1069
.
439Penpargkul
S
, Fein
F
, Sonnenblick
EH
, Scheuer
J.
Depressed cardiac sarcoplasmic reticular function from diabetic rats
.
J Mol Cell Cardiol
1981
;
13
:
303
–
309
.
440Lopaschuk
GD
, Tahiliani
AG
, Vadlamudi
RV
, Katz
S
, McNeill
JH.
Cardiac sarcoplasmic reticulum function in insulin- or carnitine-treated diabetic rats
.
Am J Physiol
1983
;
245
:
H969
–
976
.
441Lopaschuk
GD
, Katz
S
, McNeill
JH.
The effect of alloxan- and streptozotocin-induced diabetes on calcium transport in rat cardiac sarcoplasmic reticulum. The possible involvement of long chain acylcarnitines
.
Can J Physiol Pharmacol
1983
;
61
:
439
–
448
.
442Fauconnier
J
, Lanner
JT
, Zhang
SJ
, Tavi
P
, Bruton
JD
, Katz
A
, Westerblad
H.
Insulin and inositol 1,4,5-trisphosphate trigger abnormal cytosolic Ca2+ transients and reveal mitochondrial Ca2+ handling defects in cardiomyocytes of ob/ob mice
.
Diabetes
2005
;
54
:
2375
–
2381
.
443Li
SY
, Yang
X
, Ceylan-Isik
AF
, Du
M
, Sreejayan
N
, Ren
J.
Cardiac contractile dysfunction in Lep/Lep obesity is accompanied by NADPH oxidase activation, oxidative modification of sarco(endo)plasmic reticulum Ca2+-ATPase and myosin heavy chain isozyme switch
.
Diabetologia
2006
;
49
:
1434
–
1446
.
444Quan
C
, Du
Q
, Li
M
, Wang
R
, Ouyang
Q
, Su
S
, Zhu
S
, Chen
Q
, Sheng
Y
, Chen
L
, Wang
H
, Campbell
DG
, MacKintosh
C
, Yang
Z
, Ouyang
K
, Wang
HY
, Chen
S.
A PKB-SPEG signaling nexus links insulin resistance with diabetic cardiomyopathy by regulating calcium homeostasis
.
Nat Commun
2020
;
11
:
2186
.
445Shah
S
, Akhtar
MS
, Hassan
MQ
, Akhtar
M
, Paudel
YN
, Najmi
AK.
EGFR tyrosine kinase inhibition decreases cardiac remodeling and SERCA2a/NCX1 depletion in streptozotocin induced cardiomyopathy in C57/BL6 mice
.
Life Sci
2018
;
210
:
29
–
39
.
446Akhtar
MS
, Pillai
KK
, Hassan
MQ
, Dhyani
N
, Ismail
MV
, Najmi
AK.
Levosimendan reduces myocardial damage and improves cardiodynamics in streptozotocin induced diabetic cardiomyopathy via SERCA2a/NCX1 pathway
.
Life Sci
2016
;
153
:
55
–
65
.
447Fein
FS
, Kornstein
LB
, Strobeck
JE
, Capasso
JM
, Sonnenblick
EH.
Altered myocardial mechanics in diabetic rats
.
Circ Res
1980
;
47
:
922
–
933
.
448Regan
TJ
, Ettinger
PO
, Khan
MI
, Jesrani
MU
, Lyons
MM
, Oldewurtel
HA
, Weber
M.
Altered myocardial function and metabolism in chronic diabetes mellitus without ischemia in dogs
.
Circ Res
1974
;
35
:
222
–
237
.
449Garber
DW
, Neely
JR.
Decreased myocardial function and myosin ATPase in hearts from diabetic rats
.
Am J Physiol
1983
;
244
:
H586
–
591
.
450Garber
DW
, Everett
AW
, Neely
JR.
Cardiac function and myosin ATPase in diabetic rats treated with insulin, T3, and T4
.
Am J Physiol
1983
;
244
:
H592
–
598
.
451Malhotra
A
, Mordes
JP
, McDermott
L
, Schaible
TF.
Abnormal cardiac biochemistry in spontaneously diabetic Bio-Breeding/Worcester rat
.
Am J Physiol
1985
;
249
:
H1051
–
1055
.
452Dillmann
WH.
Diabetes mellitus induces changes in cardiac myosin of the rat
.
Diabetes
1980
;
29
:
579
–
582
.
453Dillmann
WH.
Influence of thyroid hormone administration on myosin ATPase activity and myosin isoenzyme distribution in the heart of diabetic rats
.
Metabolism
1982
;
31
:
199
–
204
.
454Akella
AB
, Ding
XL
, Cheng
R
, Gulati
J.
Diminished Ca2+ sensitivity of skinned cardiac muscle contractility coincident with troponin T-band shifts in the diabetic rat
.
Circ Res
1995
;
76
:
600
–
606
.
455Hofmann
PA
, Menon
V
, Gannaway
KF.
Effects of diabetes on isometric tension as a function of [Ca2+] and pH in rat skinned cardiac myocytes
.
Am J Physiol
1995
;
269
:
H1656
–
1663
.
456Wang
Y
, Fan
PS
, Kahaleh
B.
Association between enhanced type I collagen expression and epigenetic repression of the FLI1 gene in scleroderma fibroblasts
.
Arthritis Rheum
2006
;
54
:
2271
–
2279
.
457Singh
VP
, Le
B
, Khode
R
, Baker
KM
, Kumar
R.
Intracellular angiotensin II production in diabetic rats is correlated with cardiomyocyte apoptosis, oxidative stress, and cardiac fibrosis
.
Diabetes
2008
;
57
:
3297
–
3306
.
458Miller
JA.
Impact of hyperglycemia on the renin angiotensin system in early human type 1 diabetes mellitus
.
J Am Soc Nephrol
1999
;
10
:
1778
–
1785
.
459Verma
S
, Arikawa
E
, McNeill
JH.
Long-term endothelin receptor blockade improves cardiovascular function in diabetes
.
Am J Hypertens
2001
;
14
:
679
–
687
.
460Jia
G
, Habibi
J
, Bostick
BP
, Ma
L
, DeMarco
VG
, Aroor
AR
, Hayden
MR
, Whaley-Connell
AT
, Sowers
JR.
Uric acid promotes left ventricular diastolic dysfunction in mice fed a Western diet
.
Hypertension
2015
;
65
:
531
–
539
.
461Tate
M
, Grieve
DJ
, Ritchie
RH.
Are targeted therapies for diabetic cardiomyopathy on the horizon?
Clin Sci (Lond)
2017
;
131
:
897
–
915
.
462Pappachan
JM
, Sebastian
J
, Bino
BC
, Jayaprakash
K
, Vijayakumar
K
, Sujathan
P
, Adinegara
LA.
Cardiac autonomic neuropathy in diabetes mellitus: prevalence, risk factors and utility of corrected QT interval in the ECG for its diagnosis
.
Postgrad Med J
2008
;
84
:
205
–
210
.
463Voulgari
C
, Pagoni
S
, Vinik
A
, Poirier
P.
Exercise improves cardiac autonomic function in obesity and diabetes
.
Metabolism
2013
;
62
:
609
–
621
.
464Pant
T
, Dhanasekaran
A
, Bai
X
, Zhao
M
, Thorp
EB
, Forbess
JM
, Bosnjak
ZJ
, Ge
ZD.
Genome-wide differential expression profiling of lncRNAs and mRNAs associated with early diabetic cardiomyopathy
.
Sci Rep
2019
;
9
:
15345
.
465Chavali
V
, Tyagi
SC
, Mishra
PK.
Differential expression of dicer, miRNAs, and inflammatory markers in diabetic Ins2+/- Akita hearts
.
Cell Biochem Biophys
2014
;
68
:
25
–
35
.
466Dillmann
WH.
Diabetic cardiomyopathy
.
Circ Res
2019
;
124
:
1160
–
1162
.
467Karakikes
I
, Kim
M
, Hadri
L
, Sakata
S
, Sun
Y
, Zhang
W
, Chemaly
ER
, Hajjar
RJ
, Lebeche
D.
Gene remodeling in type 2 diabetic cardiomyopathy and its phenotypic rescue with SERCA2a
.
PLoS One
2009
;
4
:
e6474
.
468Li
N
, Wu
H
, Geng
R
, Tang
Q.
Identification of core gene biomarkers in patients with diabetic cardiomyopathy
.
Dis Markers
2018
;
2018
:
6025061
.
469Gollmer
J
, Zirlik
A
, Bugger
H.
Mitochondrial mechanisms in diabetic cardiomyopathy
.
Diabetes Metab J
2020
;
44
:
33
–
53
.
470Hu
L
, Ding
M
, Tang
D
, Gao
E
, Li
C
, Wang
K
, Qi
B
, Qiu
J
, Zhao
H
, Chang
P
, Fu
F
, Li
Y.
Targeting mitochondrial dynamics by regulating Mfn2 for therapeutic intervention in diabetic cardiomyopathy
.
Theranostics
2019
;
9
:
3687
–
3706
.
471Duncan
JG
, Fong
JL
, Medeiros
DM
, Finck
BN
, Kelly
DP.
Insulin-resistant heart exhibits a mitochondrial biogenic response driven by the peroxisome proliferator-activated receptor-alpha/PGC-1alpha gene regulatory pathway
.
Circulation
2007
;
115
:
909
–
917
.
472Beermann
J
, Piccoli
MT
, Viereck
J
, Thum
T.
Non-coding RNAs in development and disease: background, mechanisms, and therapeutic approaches
.
Physiol Rev
2016
;
96
:
1297
–
1325
.
473Seal
RL
, Chen
LL
, Griffiths-Jones
S
, Lowe
TM
, Mathews
MB
, O'Reilly
D
, Pierce
AJ
, Stadler
PF
, Ulitsky
I
, Wolin
SL
, Bruford
EA.
A guide to naming human non-coding RNA genes
.
EMBO J
2020
;
39
:
e103777
.
474Zhang
W
, Xu
W
, Feng
Y
, Zhou
X.
Non-coding RNA involvement in the pathogenesis of diabetic cardiomyopathy
.
J Cell Mol Med
2019
;
23
:
5859
–
5867
.
475de Gonzalo-Calvo
D
, Kenneweg
F
, Bang
C
, Toro
R
, van der Meer
RW
, Rijzewijk
LJ
, Smit
JW
, Lamb
HJ
, Llorente-Cortes
V
, Thum
T.
Circulating long-non coding RNAs as biomarkers of left ventricular diastolic function and remodelling in patients with well-controlled type 2 diabetes
.
Sci Rep
2016
;
6
:
37354
.
476Li
D
, Kular
L
, Vij
M
, Herter
EK
, Li
X
, Wang
A
, Chu
T
, Toma
M-A
, Zhang
L
, Liapi
E
, Mota
A
, Blomqvist
L
, Gallais Sérézal
I
, Rollman
O
, Wikstrom
JD
, Bienko
M
, Berglund
D
, Ståhle
M
, Sommar
P
, Jagodic
M
, Landén
NX.
Human skin long noncoding RNA WAKMAR1 regulates wound healing by enhancing keratinocyte migration
.
Proc Natl Acad Sci USA
2019
;
116
:
9443
–
9452
.
477Wang
P
, Xue
Y
, Han
Y
, Lin
L
, Wu
C
, Xu
S
, Jiang
Z
, Xu
J
, Liu
Q
, Cao
X.
The STAT3-binding long noncoding RNA lnc-DC controls human dendritic cell differentiation
.
Science
2014
;
344
:
310
–
313
.
478Gao
L
, Wang
XF
, Guo
S
, Xiao
LL
, Liang
C
, Wang
Z
, Li
YP
, Liu
YZ
, Yao
R
, Liu
Y
, Zhang
YZ.
LncRNA HOTAIR functions as a competing endogenous RNA to upregulate SIRT1 by sponging miR-34a in diabetic cardiomyopathy
.
J Cell Physiol
2019
;
234
:
4944
–
4958
.
479Zhou
X
, Zhang
W
, Jin
MC
, Chen
JC
, Xu
WT
, Kong
XQ.
lncRNA MIAT functions as a competing endogenous RNA to upregulate DAPK2 by sponging miR-22-3p in diabetic cardiomyopathy
.
Cell Death Dis
2017
;
8
:
e2929
.
480Zhuo
C
, Jiang
R
, Lin
X
, Shao
M.
LncRNA H19 inhibits autophagy by epigenetically silencing of DIRAS3 in diabetic cardiomyopathy
.
Oncotarget
2017
;
8
:
1429
–
1437
.
481Feng
B
, Chen
S
, Chakrabarti
S.
Long noncoding RNA Zfas1 in diabetic cardiomyopathy
.
Am Diabetes Assoc
2018
;
67
:
473-P
.
482Li
X
, Wang
H
, Yao
B
, Xu
W
, Chen
J
, Zhou
X.
lncRNA H19/miR-675 axis regulates cardiomyocyte apoptosis by targeting VDAC1 in diabetic cardiomyopathy
.
Sci Rep
2016
;
6
:
36340
.
483Raut
SK
, Singh
GB
, Rastogi
B
, Saikia
UN
, Mittal
A
, Dogra
N
, Singh
S
, Prasad
R
, Khullar
M.
miR-30c and miR-181a synergistically modulate p53-p21 pathway in diabetes induced cardiac hypertrophy
.
Mol Cell Biochem
2016
;
417
:
191
–
203
.
484Feng
B
, Chen
S
, George
B
, Feng
Q
, Chakrabarti
S.
miR133a regulates cardiomyocyte hypertrophy in diabetes
.
Diabetes Metab Res Rev
2010
;
26
:
40
–
49
.
485Duan
Y
, Zhou
B
, Su
H
, Liu
Y
, Du
C.
miR-150 regulates high glucose-induced cardiomyocyte hypertrophy by targeting the transcriptional co-activator p300
.
Exp Cell Res
2013
;
319
:
173
–
184
.
486Shen
E
, Diao
X
, Wang
X
, Chen
R
, Hu
B.
MicroRNAs involved in the mitogen-activated protein kinase cascades pathway during glucose-induced cardiomyocyte hypertrophy
.
Am J Pathol
2011
;
179
:
639
–
650
.
487Kuwabara
Y
, Horie
T
, Baba
O
, Watanabe
S
, Nishiga
M
, Usami
S
, Izuhara
M
, Nakao
T
, Nishino
T
, Otsu
K
, Kita
T
, Kimura
T
, Ono
K.
MicroRNA-451 exacerbates lipotoxicity in cardiac myocytes and high-fat diet-induced cardiac hypertrophy in mice through suppression of the LKB1/AMPK pathway
.
Circ Res
2015
;
116
:
279
–
U217
.
488Chen
S
, Puthanveetil
P
, Feng
B
, Matkovich
SJ
, Dorn
GW
2nd, Chakrabarti
S.
Cardiac miR-133a overexpression prevents early cardiac fibrosis in diabetes
.
J Cell Mol Med
2014
;
18
:
415
–
421
.
489Rawal
S
, Munasinghe
PE
, Nagesh
PT
, Lew
JKS
, Jones
GT
, Williams
MJA
, Davis
P
, Bunton
D
, Galvin
IF
, Manning
P
, Lamberts
RR
, Katare
R.
Down-regulation of miR-15a/b accelerates fibrotic remodelling in the Type 2 diabetic human and mouse heart
.
Clin Sci (Lond)
2017
;
131
:
847
–
863
.
490Feng
B
, Chen
S
, Gordon
AD
, Chakrabarti
S.
miR-146a mediates inflammatory changes and fibrosis in the heart in diabetes
.
J Mol Cell Cardiol
2017
;
105
:
70
–
76
.
491Moore
A
, Shindikar
A
, Fomison-Nurse
I
, Riu
F
, Munasinghe
PE
, Ram
TP
, Saxena
P
, Coffey
S
, Bunton
RW
, Galvin
IF
, Williams
MJ
, Emanueli
C
, Madeddu
P
, Katare
R.
Rapid onset of cardiomyopathy in STZ-induced female diabetic mice involves the downregulation of pro-survival Pim-1
.
Cardiovasc Diabetol
2014
;
13
:
68
.
492Zheng
D
, Ma
J
, Yu
Y
, Li
M
, Ni
R
, Wang
G
, Chen
R
, Li
J
, Fan
GC
, Lacefield
JC
, Peng
T.
Silencing of miR-195 reduces diabetic cardiomyopathy in C57BL/6 mice
.
Diabetologia
2015
;
58
:
1949
–
1958
.
493Qiao
Y
, Zhao
Y
, Liu
Y
, Ma
N
, Wang
C
, Zou
J
, Liu
Z
, Zhou
Z
, Han
D
, He
J
, Sun
Q
, Liu
Y
, Xu
C
, Du
Z
, Huang
H.
miR-483-3p regulates hyperglycaemia-induced cardiomyocyte apoptosis in transgenic mice
.
Biochem Biophys Res Commun
2016
;
477
:
541
–
547
.
494Arnold
N
, Koppula
PR
, Gul
R
, Luck
C
, Pulakat
L.
Regulation of cardiac expression of the diabetic marker microRNA miR-29
.
PLoS One
2014
;
9
:
e103284
.
495Yildirim
SS
, Akman
D
, Catalucci
D
, Turan
B.
Relationship between downregulation of miRNAs and increase of oxidative stress in the development of diabetic cardiac dysfunction: junctin as a target protein of miR-1
.
Cell Biochem Biophys
2013
;
67
:
1397
–
1408
.
496Yu
M
, Liu
Y
, Zhang
B
, Shi
Y
, Cui
L
, Zhao
X.
Inhibiting microRNA-144 abates oxidative stress and reduces apoptosis in hearts of streptozotocin-induced diabetic mice
.
Cardiovasc Pathol
2015
;
24
:
375
–
381
.
497Reddy
MA
, Jin
W
, Villeneuve
L
, Wang
M
, Lanting
L
, Todorov
I
, Kato
M
, Natarajan
R.
Pro-inflammatory role of microrna-200 in vascular smooth muscle cells from diabetic mice
.
Arterioscler Thromb Vasc Biol
2012
;
32
:
721
–
729
.
498Ashwal-Fluss
R
, Meyer
M
, Pamudurti
NR
, Ivanov
A
, Bartok
O
, Hanan
M
, Evantal
N
, Memczak
S
, Rajewsky
N
, Kadener
S.
circRNA biogenesis competes with pre-mRNA splicing
.
Mol Cell
2014
;
56
:
55
–
66
.
499Li
Z
, Huang
C
, Bao
C
, Chen
L
, Lin
M
, Wang
X
, Zhong
G
, Yu
B
, Hu
W
, Dai
L
, Zhu
P
, Chang
Z
, Wu
Q
, Zhao
Y
, Jia
Y
, Xu
P
, Liu
H
, Shan
G.
Exon-intron circular RNAs regulate transcription in the nucleus
.
Nat Struct Mol Biol
2015
;
22
:
256
–
264
.
500Dong
SZ
, Tu
CY
, Ye
X
, Li
LL
, Zhang
MC
, Xue
AM
, Chen
SH
, Zhao
ZQ
, Cong
B
, Lin
JY
, Shen
YW.
Expression profiling of circular RNAs and their potential role in early-stage diabetic cardiomyopathy
.
Mol Med Rep
2020
;
22
:
1958
–
1968
.
501Tang
N
, Jiang
S
, Yang
Y
, Liu
S
, Ponnusamy
M
, Xin
H
, Yu
T.
Noncoding RNAs as therapeutic targets in atherosclerosis with diabetes mellitus
.
Cardiovasc Ther
2018
;
36
:
e12436
.
502Zhou
B
, Yu
JW.
A novel identified circular RNA, circRNA_010567, promotes myocardial fibrosis via suppressing miR-141 by targeting TGF-beta1
.
Biochem Biophys Res Commun
2017
;
487
:
769
–
775
.
503Wang
W
, Zhang
S
, Xu
L
, Feng
Y
, Wu
X
, Zhang
M
, Yu
Z
, Zhou
X.
Involvement of circHIPK3 in the pathogenesis of diabetic cardiomyopathy in mice
.
Diabetologia
2021
;
64
:
681
–
692
.
504Yang
F
, Li
A
, Qin
Y
, Che
H
, Wang
Y
, Lv
J
, Li
Y
, Li
H
, Yue
E
, Ding
X
, Yu
Y
, Bai
Y
, Wang
L.
A novel circular RNA mediates pyroptosis of diabetic cardiomyopathy by functioning as a competing endogenous RNA
.
Mol Ther Nucleic Acids
2019
;
17
:
636
–
643
.
505Gaikwad
AB
, Sayyed
SG
, Lichtnekert
J
, Tikoo
K
, Anders
HJ.
Renal failure increases cardiac histone h3 acetylation, dimethylation, and phosphorylation and the induction of cardiomyopathy-related genes in type 2 diabetes
.
Am J Pathol
2010
;
176
:
1079
–
1083
.
506Miao
F
, Gonzalo
IG
, Lanting
L
, Natarajan
R.
In vivo chromatin remodeling events leading to inflammatory gene transcription under diabetic conditions
.
J Biol Chem
2004
;
279
:
18091
–
18097
.
507Malek
V
, Sharma
N
, Gaikwad
AB.
Histone acetylation regulates natriuretic peptides and neprilysin gene expressions in diabetic cardiomyopathy and nephropathy
.
Curr Mol Pharmacol
2019
;
12
:
61
–
71
.
508Uchihashi
M
, Hoshino
A
, Okawa
Y
, Ariyoshi
M
, Kaimoto
S
, Tateishi
S
, Ono
K
, Yamanaka
R
, Hato
D
, Fushimura
Y
, Honda
S
, Fukai
K
, Higuchi
Y
, Ogata
T
, Iwai-Kanai
E
, Matoba
S.
Cardiac-specific Bdh1 overexpression ameliorates oxidative stress and cardiac remodeling in pressure overload-induced heart failure
.
Circ Heart Fail
2017
;
10
:
e004417
.
509van den Berge
JC
, Constantinescu
AA
, Boiten
HJ
, van Domburg
RT
, Deckers
JW
, Akkerhuis
KM.
Short- and long-term prognosis of patients with acute heart failure with and without diabetes: changes over the last three decades
.
Diabetes Care
2018
;
41
:
143
–
149
.
510Kenny
HC
, Abel
ED.
Heart failure in type 2 diabetes mellitus
.
Circ Res
2019
;
124
:
121
–
141
.
511Cameron
AR
, Morrison
VL
, Levin
D
, Mohan
M
, Forteath
C
, Beall
C
, McNeilly
AD
, Balfour
DJ
, Savinko
T
, Wong
AK
, Viollet
B
, Sakamoto
K
, Fagerholm
SC
, Foretz
M
, Lang
CC
, Rena
G.
Anti-inflammatory effects of metformin irrespective of diabetes status
.
Circ Res
2016
;
119
:
652
–
665
.
512Dziubak
A
, Wójcicka
G
, Wojtak
A
, Bełtowski
J.
Metabolic effects of metformin in the failing heart
.
Int J Mol Sci
2018
;
19
:
2869
.
513El Messaoudi
S
, Rongen
GA
, de Boer
RA
, Riksen
NP.
The cardioprotective effects of metformin
.
Curr Opin Lipidol
2011
;
22
:
445
–
453
.
514Eurich
DT
, Tsuyuki
RT
, Majumdar
SR
, McAlister
FA
, Lewanczuk
R
, Shibata
MC
, Johnson
JA.
Metformin treatment in diabetes and heart failure: when academic equipoise meets clinical reality
.
Trials
2009
;
10
:
1745
–
6215
.
515Dludla
PV
, Nyambuya
TM
, Johnson
R
, Silvestri
S
, Orlando
P
, Mazibuko-Mbeje
SE
, Gabuza
KB
, Mxinwa
V
, Mokgalaboni
K
, Tiano
L
, Muller
CJF
, Louw
J
, Nkambule
BB.
Metformin and heart failure-related outcomes in patients with or without diabetes: a systematic review of randomized controlled trials
.
Heart Fail Rev
2020
; DOI: 10.1007/s10741-020-09942-y.
516Weir
DL
, Abrahamowicz
M
, Beauchamp
ME
, Eurich
DT.
Acute vs cumulative benefits of metformin use in patients with type 2 diabetes and heart failure
.
Diabetes Obes Metab
2018
;
20
:
2653
–
2660
.
517Retwiński
A
, Kosmalski
M
, Crespo-Leiro
M
, Maggioni
A
, Opolski
G
, Ponikowski
P
, Poloński
L
, Jankowska
E
, Drzewoski
J
, Drożdż
J.
The influence of metformin and the presence of type 2 diabetes mellitus on mortality and hospitalisation in patients with heart failure
.
Kardiol Pol
2018
;
76
:
1336
–
1343
.
518Pantalone
KM
, Kattan
MW
, Yu
C
, Wells
BJ
, Arrigain
S
, Jain
A
, Atreja
A
, Zimmerman
RS.
The risk of developing coronary artery disease or congestive heart failure, and overall mortality, in type 2 diabetic patients receiving rosiglitazone, pioglitazone, metformin, or sulfonylureas: a retrospective analysis
.
Acta Diabetol
2009
;
46
:
145
–
154
.
519Sola
D
, Rossi
L
, Schianca
GP
, Maffioli
P
, Bigliocca
M
, Mella
R
, Corliano
F
, Fra
GP
, Bartoli
E
, Derosa
G.
Sulfonylureas and their use in clinical practice
.
Arch Med Sci
2015
;
11
:
840
–
848
.
520Monami
M
, Genovese
S
, Mannucci
E.
Cardiovascular safety of sulfonylureas: a meta-analysis of randomized clinical trials
.
Diabetes Obes Metab
2013
;
15
:
938
–
953
.
521Rao
AD
, Kuhadiya
N
, Reynolds
K
, Fonseca
VA.
Is the combination of sulfonylureas and metformin associated with an increased risk of cardiovascular disease or all-cause mortality?: a meta-analysis of observational studies
.
Diabetes Care
2008
;
31
:
1672
–
1678
.
522Roumie
CL
, Greevy
RA
, Grijalva
CG
, Hung
AM
, Liu
X
, Murff
HJ
, Elasy
TA
, Griffin
MR.
Association between intensification of metformin treatment with insulin vs sulfonylureas and cardiovascular events and all-cause mortality among patients with diabetes
.
JAMA
2014
;
311
:
2288
–
2296
.
523Evans
JM
, Ogston
SA
, Emslie-Smith
A
, Morris
AD.
Risk of mortality and adverse cardiovascular outcomes in type 2 diabetes: a comparison of patients treated with sulfonylureas and metformin
.
Diabetologia
2006
;
49
:
930
–
936
.
524Azoulay
L
, Suissa
S.
Sulfonylureas and the risks of cardiovascular events and death: a methodological meta-regression analysis of the observational studies
.
Diabetes Care
2017
;
40
:
706
–
714
.
525Simpson
SH
, Lee
J
, Choi
S
, Vandermeer
B
, Abdelmoneim
AS
, Featherstone
TR.
Mortality risk among sulfonylureas: a systematic review and network meta-analysis
.
Lancet Diabetes Endocrinol
2015
;
3
:
43
–
51
.
526Douros
A
, Yin
H
, Yu
OHY
, Filion
KB
, Azoulay
L
, Suissa
S.
Pharmacologic differences of sulfonylureas and the risk of adverse cardiovascular and hypoglycemic events
.
Diabetes Care
2017
;
40
:
1506
–
1513
.
527Diabetes Canada Clinical Practice Guidelines Expert
C
, Connelly
KA
, Gilbert
RE
, Liu
P
; Diabetes Canada Clinical Practice Guidelines Expert Committee.
Treatment of diabetes in people with heart failure
.
Can J Diabetes
2018
;
42(Suppl. 1
):
S196
–
S200
.
528Charbonnel
B
, Dormandy
J
, Erdmann
E
, Massi-Benedetti
M
, Skene
A
; PROactive Study Group.
The prospective pioglitazone clinical trial in macrovascular events (PROactive): can pioglitazone reduce cardiovascular events in diabetes? Study design and baseline characteristics of 5238 patients
.
Diabetes Care
2004
;
27
:
1647
–
1653
.
529Investigators
DT
, Dagenais
GR
, Gerstein
HC
, Holman
R
, Budaj
A
, Escalante
A
, Hedner
T
, Keltai
M
, Lonn
E
, McFarlane
S
, McQueen
M
, Teo
K
, Sheridan
P
, Bosch
J
, Pogue
J
, Yusuf
S.
Effects of ramipril and rosiglitazone on cardiovascular and renal outcomes in people with impaired glucose tolerance or impaired fasting glucose: results of the Diabetes REduction Assessment with ramipril and rosiglitazone Medication (DREAM) trial
.
Diabetes Care
2008
;
31
:
1007
–
1014
.
530Home
PD
, Pocock
SJ
, Beck-Nielsen
H
, Curtis
PS
, Gomis
R
, Hanefeld
M
, Jones
NP
, Komajda
M
, McMurray
JJ
; RECORD Study Team.
Rosiglitazone evaluated for cardiovascular outcomes in oral agent combination therapy for type 2 diabetes (RECORD): a multicentre, randomised, open-label trial
.
Lancet
2009
;
373
:
2125
–
2135
.
531Kendall
DM
, Rubin
CJ
, Mohideen
P
, Ledeine
JM
, Belder
R
, Gross
J
, Norwood
P
, O'Mahony
M
, Sall
K
, Sloan
G
, Roberts
A
, Fiedorek
FT
, DeFronzo
RA.
Improvement of glycemic control, triglycerides, and HDL cholesterol levels with muraglitazar, a dual (alpha/gamma) peroxisome proliferator-activated receptor activator, in patients with type 2 diabetes inadequately controlled with metformin monotherapy: a double-blind, randomized, pioglitazone-comparative study
.
Diabetes Care
2006
;
29
:
1016
–
1023
.
532Kalliora
C
, Drosatos
K.
The glitazars paradox: cardiotoxicity of the metabolically beneficial dual PPARalpha and PPARgamma activation
.
J Cardiovasc Pharmacol
2020
;
76
:
514
–
526
.
533Nissen
SE
, Wolski
K
, Topol
EJ.
Effect of muraglitazar on death and major adverse cardiovascular events in patients with type 2 diabetes mellitus
.
JAMA
2005
;
294
:
2581
–
2586
.
534Lincoff
AM
, Tardif
JC
, Schwartz
GG
, Nicholls
SJ
, Ryden
L
, Neal
B
, Malmberg
K
, Wedel
H
, Buse
JB
, Henry
RR
, Weichert
A
, Cannata
R
, Svensson
A
, Volz
D
, Grobbee
DE
, AleCardio
I.
Effect of aleglitazar on cardiovascular outcomes after acute coronary syndrome in patients with type 2 diabetes mellitus: the AleCardio randomized clinical trial
.
JAMA
2014
;
311
:
1515
–
1525
.
535Hui
H
, Farilla
L
, Merkel
P
, Perfetti
R.
The short half-life of glucagon-like peptide-1 in plasma does not reflect its long-lasting beneficial effects
.
Eur J Endocrinol
2002
;
146
:
863
–
869
.
536Rachman
J
, Barrow
BA
, Levy
JC
, Turner
RC.
Near-normalisation of diurnal glucose concentrations by continuous administration of glucagon-like peptide-1 (GLP-1) in subjects with NIDDM
.
Diabetologia
1997
;
40
:
205
–
211
.
537Mojsov
S
, Weir
G
, Habener
J.
Insulinotropin: glucagon-like peptide I (7-37) co-encoded in the glucagon gene is a potent stimulator of insulin release in the perfused rat pancreas
.
J Clin Invest
1987
;
79
:
616
–
619
.
538Ussher
JR
, Drucker
DJ.
Cardiovascular biology of the incretin system
.
Endocr Rev
2012
;
33
:
187
–
215
.
539Marso
SP
, Bain
SC
, Consoli
A
, Eliaschewitz
FG
, Jodar
E
, Leiter
LA
, Lingvay
I
, Rosenstock
J
, Seufert
J
, Warren
ML
, Woo
V
, Hansen
O
, Holst
AG
, Pettersson
J
, Vilsboll
T
; SUSTAIN-6 Investigators.
Semaglutide and cardiovascular outcomes in patients with type 2 diabetes
.
N Engl J Med
2016
;
375
:
1834
–
1844
.
540Holman
RR
, Bethel
MA
, Mentz
RJ
, Thompson
VP
, Lokhnygina
Y
, Buse
JB
, Chan
JC
, Choi
J
, Gustavson
SM
, Iqbal
N
, Maggioni
AP
, Marso
SP
, Ohman
P
, Pagidipati
NJ
, Poulter
N
, Ramachandran
A
, Zinman
B
, Hernandez
AF
; EXSCEL Study Group.
Effects of once-weekly exenatide on cardiovascular outcomes in type 2 diabetes
.
N Engl J Med
2017
;
377
:
1228
–
1239
.
541Pfeffer
MA
, Claggett
B
, Diaz
R
, Dickstein
K
, Gerstein
HC
, Kober
LV
, Lawson
FC
, Ping
L
, Wei
X
, Lewis
EF
, Maggioni
AP
, McMurray
JJ
, Probstfield
JL
, Riddle
MC
, Solomon
SD
, Tardif
JC
; ELIXA Investigators.
Lixisenatide in patients with type 2 diabetes and acute coronary syndrome
.
N Engl J Med
2015
;
373
:
2247
–
2257
.
542Marso
SP
, Daniels
GH
, Brown-Frandsen
K
, Kristensen
P
, Mann
JF
, Nauck
MA
, Nissen
SE
, Pocock
S
, Poulter
NR
, Ravn
LS
, Steinberg
WM
, Stockner
M
, Zinman
B
, Bergenstal
RM
, Buse
JB
; LEADER Steering Committee, LEADER Trial Investigators.
Liraglutide and cardiovascular outcomes in type 2 diabetes
.
N Engl J Med
2016
;
375
:
311
–
322
.
543Margulies
KB
, Hernandez
AF
, Redfield
MM
, Givertz
MM
, Oliveira
GH
, Cole
R
, Mann
DL
, Whellan
DJ
, Kiernan
MS
, Felker
GM
, McNulty
SE
, Anstrom
KJ
, Shah
MR
, Braunwald
E
, Cappola
TP;
NHLBI Heart Failure Clinical Research Network.
Effects of liraglutide on clinical stability among patients with advanced heart failure and reduced ejection fraction a randomized clinical trial
.
J Am Med Assoc
2016
;
316
:
500
–
508
.
544Jorsal
A
, Kistorp
C
, Holmager
P
, Tougaard
RS
, Nielsen
R
, Hänselmann
A
, Nilsson
B
, Møller
JE
, Hjort
J
, Rasmussen
J
, Boesgaard
TW
, Schou
M
, Videbæk
L
, Gustafsson
I
, Flyvbjerg
A
, Wiggers
H
, Tarnow
L.
Effect of liraglutide, a glucagon‐like peptide‐1 analogue, on left ventricular function in stable chronic heart failure patients with and without diabetes (LIVE)—a multicentre, double‐blind, randomised, placebo‐controlled trial
.
Eur J Heart Fail
2017
;
19
:
69
–
77
.
545Mentlein
R
, Gallwitz
B
, Schmidt
WE.
Dipeptidyl‐peptidase IV hydrolyses gastric inhibitory polypeptide, glucagon‐like peptide‐1 (7–36) amide, peptide histidine methionine and is responsible for their degradation in human serum
.
Eur J Biochem
1993
;
214
:
829
–
835
.
546Deacon
CF
, Hughes
TE
, Holst
JJ.
Dipeptidyl peptidase IV inhibition potentiates the insulinotropic effect of glucagon-like peptide 1 in the anesthetized pig
.
Diabetes
1998
;
47
:
764
–
769
.
547Ahren
B
, Holst
JJ
, Martensson
H
, Balkan
B.
Improved glucose tolerance and insulin secretion by inhibition of dipeptidyl peptidase IV in mice
.
Eur J Pharmacol
2000
;
404
:
239
–
245
.
548Gutzwiller
JP
, Goke
B
, Drewe
J
, Hildebrand
P
, Ketterer
S
, Handschin
D
, Winterhalder
R
, Conen
D
, Beglinger
C.
Glucagon-like peptide-1: a potent regulator of food intake in humans
.
Gut
1999
;
44
:
81
–
86
.
549Dicker
D.
DPP-4 inhibitors: impact on glycemic control and cardiovascular risk factors
.
Diabetes Care
2011
;
34(Suppl. 2
):
S276
–
278
.
550Burkey
BF
, Li
X
, Bolognese
L
, Balkan
B
, Mone
M
, Russell
M
, Hughes
TE
, Wang
PR.
Acute and chronic effects of the incretin enhancer vildagliptin in insulin-resistant rats
.
J Pharmacol Exp Ther
2005
;
315
:
688
–
695
.
551Ahren
B
, Simonsson
E
, Larsson
H
, Landin-Olsson
M
, Torgeirsson
H
, Jansson
PA
, Sandqvist
M
, Bavenholm
P
, Efendic
S
, Eriksson
JW
, Dickinson
S
, Holmes
D.
Inhibition of dipeptidyl peptidase IV improves metabolic control over a 4-week study period in type 2 diabetes
.
Diabetes Care
2002
;
25
:
869
–
875
.
552Ahren
B
, Landin-Olsson
M
, Jansson
PA
, Svensson
M
, Holmes
D
, Schweizer
A.
Inhibition of dipeptidyl peptidase-4 reduces glycemia, sustains insulin levels, and reduces glucagon levels in type 2 diabetes
.
J Clin Endocrinol Metab
2004
;
89
:
2078
–
2084
.
553Ristic
S
, Byiers
S
, Foley
J
, Holmes
D.
Improved glycaemic control with dipeptidyl peptidase-4 inhibition in patients with type 2 diabetes: vildagliptin (LAF237) dose response
.
Diabetes Obes Metab
2005
;
7
:
692
–
698
.
554Scirica
BM
, Bhatt
DL
, Braunwald
E
, Steg
PG
, Davidson
J
, Hirshberg
B
, Ohman
P
, Frederich
R
, Wiviott
SD
, Hoffman
EB
, Cavender
MA
, Udell
JA
, Desai
NR
, Mosenzon
O
, McGuire
DK
, Ray
KK
, Leiter
LA
, Raz
I
, SAVOR-TIMI 53 Steering Committee and Investigators.
Saxagliptin and cardiovascular outcomes in patients with type 2 diabetes mellitus
.
N Engl J Med
2013
;
369
:
1317
–
1326
.
555White
WB
, Cannon
CP
, Heller
SR
, Nissen
SE
, Bergenstal
RM
, Bakris
GL
, Perez
AT
, Fleck
PR
, Mehta
CR
, Kupfer
S
, Wilson
C
, Cushman
WC
, Zannad
F
, Investigators
E.
Alogliptin after acute coronary syndrome in patients with type 2 diabetes
.
N Engl J Med
2013
;
369
:
1327
–
1335
.
556White
WB
, Kupfer
S
, Zannad
F
, Mehta
CR
, Wilson
CA
, Lei
L
, Bakris
GL
, Nissen
SE
, Cushman
WC
, Heller
SR
, Bergenstal
RM
, Fleck
PR
, Cannon
CP
; EXAMINE Investigators.
Cardiovascular mortality in patients with type 2 diabetes and recent acute coronary syndromes from the EXAMINE Trial
.
Diabetes Care
2016
;
39
:
1267
–
1273
.
557Green
JB
, Bethel
MA
, Armstrong
PW
, Buse
JB
, Engel
SS
, Garg
J
, Josse
R
, Kaufman
KD
, Koglin
J
, Korn
S
, Lachin
JM
, McGuire
DK
, Pencina
MJ
, Standl
E
, Stein
PP
, Suryawanshi
S
, Van de Werf
F
, Peterson
ED
, Holman
RR
; TECOS Study Group.
Effect of sitagliptin on cardiovascular outcomes in type 2 diabetes
.
N Engl J Med
2015
;
373
:
232
–
242
.
558Fisher
M.
Series: cardiovascular outcome trials for diabetes drugs Sitagliptin and TECOS
.
Br J Diabetes
2020
;
20
:
55
–
57
.
559Oyama
J
, Murohara
T
, Kitakaze
M
, Ishizu
T
, Sato
Y
, Kitagawa
K
, Kamiya
H
, Ajioka
M
, Ishihara
M
, Dai
K
, Nanasato
M
, Sata
M
, Maemura
K
, Tomiyama
H
, Higashi
Y
, Kaku
K
, Yamada
H
, Matsuhisa
M
, Yamashita
K
, Bando
YK
, Kashihara
N
, Ueda
S
, Inoue
T
, Tanaka
A
, Node
K
; PROLOGUE Study Investigators
The effect of sitagliptin on carotid artery atherosclerosis in type 2 diabetes: the PROLOGUE randomized controlled trial
.
PLoS Med
2016
;
13
:
e1002051
.
560Ou
SM
, Chen
HT
, Kuo
SC
, Chen
TJ
, Shih
CJ
, Chen
YT.
Dipeptidyl peptidase-4 inhibitors and cardiovascular risks in patients with pre-existing heart failure
.
Heart
2017
;
103
:
414
–
420
.
561Sato
A
, Yoshihisa
A
, Kanno
Y
, Takiguchi
M
, Miura
S
, Shimizu
T
, Nakamura
Y
, Yamauchi
H
, Owada
T
, Sato
T
, Suzuki
S
, Oikawa
M
, Yamaki
T
, Sugimoto
K
, Kunii
H
, Nakazato
K
, Suzuki
H
, Saitoh
SI
, Takeishi
Y.
Associations of dipeptidyl peptidase-4 inhibitors with mortality in hospitalized heart failure patients with diabetes mellitus
.
ESC Heart Fail
2016
;
3
:
77
–
85
.
562Giorda
CB
, Picariello
R
, Tartaglino
B
, Marafetti
L
, Di Noi
F
, Alessiato
A
, Costa
G
, Gnavi
R.
Hospitalisation for heart failure and mortality associated with dipeptidyl peptidase 4 (DPP-4) inhibitor use in an unselected population of subjects with type 2 diabetes: a nested case-control study
.
Bmj Open
2015
;
5
:
e007959
.
563Monami
M
, Dicembrini
I
, Martelli
D
, Mannucci
E.
Safety of dipeptidyl peptidase-4 inhibitors: a meta-analysis of randomized clinical trials
.
Curr Med Res Opin
2011
;
27(Suppl. 3
):
57
–
64
.
564Clifton
P.
Do dipeptidyl peptidase IV (DPP-IV) inhibitors cause heart failure?
Clin Ther
2014
;
36
:
2072
–
2079
.
565Wu
S
, Hopper
I
, Skiba
M
, Krum
H.
Dipeptidyl peptidase-4 inhibitors and cardiovascular outcomes: meta-analysis of randomized clinical trials with 55,141 participants
.
Cardiovasc Ther
2014
;
32
:
147
–
158
.
566Hamdani
N
, Hervent
AS
, Vandekerckhove
L
, Matheeussen
V
, Demolder
M
, Baerts
L
, De Meester
I
, Linke
WA
, Paulus
WJ
, De Keulenaer
G.
Left ventricular diastolic dysfunction and myocardial stiffness in diabetic mice is attenuated by inhibition of dipeptidyl peptidase 4
.
Cardiovasc Res
2014
;
104
:
423
–
431
.
567Yin
M
, Sillje
HH
, Meissner
M
, van Gilst
WH
, de Boer
RA.
Early and late effects of the DPP-4 inhibitor vildagliptin in a rat model of post-myocardial infarction heart failure
.
Cardiovasc Diabetol
2011
;
10
:
85
.
568Gallo
LA
, Wright
EM
, Vallon
V.
Probing SGLT2 as a therapeutic target for diabetes: basic physiology and consequences
.
Diab Vasc Dis Res
2015
;
12
:
78
–
89
.
569Hattersley
AT
, Thorens
B.
Type 2 diabetes, SGLT2 inhibitors, and glucose secretion
.
N Engl J Med
2015
;
373
:
974
–
976
.
570Vallon
V
, Thomson
SC.
Targeting renal glucose reabsorption to treat hyperglycaemia: the pleiotropic effects of SGLT2 inhibition
.
Diabetologia
2017
;
60
:
215
–
225
.
571Zinman
B
, Wanner
C
, Lachin
JM
, Fitchett
D
, Bluhmki
E
, Hantel
S
, Mattheus
M
, Devins
T
, Johansen
OE
, Woerle
HJ
, Broedl
UC
, Inzucchi
SE;
EMPA-REG OUTCOME Investigators.
Empagliflozin, cardiovascular outcomes, and mortality in type 2 diabetes
.
N Engl J Med
2015
;
373
:
2117
–
2128
.
572Radholm
K
, Figtree
G
, Perkovic
V
, Solomon
SD
, Mahaffey
KW
, de Zeeuw
D
, Fulcher
G
, Barrett
TD
, Shaw
W
, Desai
M
, Matthews
DR
, Neal
B.
Canagliflozin and heart failure in type 2 diabetes mellitus: results from the CANVAS program
.
Circulation
2018
;
138
:
458
–
468
.
573McMurray
JJV
, DeMets
DL
, Inzucchi
SE
, Kober
L
, Kosiborod
MN
, Langkilde
AM
, Martinez
FA
, Bengtsson
O
, Ponikowski
P
, Sabatine
MS
, Sjostrand
M
, Solomon
SD
; DAPA-HF Committees and Investigators.
A trial to evaluate the effect of the sodium-glucose co-transporter 2 inhibitor dapagliflozin on morbidity and mortality in patients with heart failure and reduced left ventricular ejection fraction (DAPA-HF)
.
Eur J Heart Fail
2019
;
21
:
665
–
675
.
574Packer
M
, Anker
SD
, Butler
J
, Filippatos
G
, Pocock
SJ
, Carson
P
, Januzzi
J
, Verma
S
, Tsutsui
H
, Brueckmann
M
, Jamal
W
, Kimura
K
, Schnee
J
, Zeller
C
, Cotton
D
, Bocchi
E
, Bohm
M
, Choi
DJ
, Chopra
V
, Chuquiure
E
, Giannetti
N
, Janssens
S
, Zhang
J
, Gonzalez Juanatey
JR
, Kaul
S
, Brunner-La Rocca
HP
, Merkely
B
, Nicholls
SJ
, Perrone
S
, Pina
I
, Ponikowski
P
, Sattar
N
, Senni
M
, Seronde
MF
, Spinar
J
, Squire
I
, Taddei
S
, Wanner
C
, Zannad
F
; EMPEROR-Reduced Trial Investigators.
Cardiovascular and renal outcomes with empagliflozin in heart failure
.
N Engl J Med
2020
;
383
:
1413
–
1424
.
575Wiviott
SD
, Raz
I
, Bonaca
MP
, Mosenzon
O
, Kato
ET
, Cahn
A
, Silverman
MG
, Zelniker
TA
, Kuder
JF
, Murphy
SA
, Bhatt
DL
, Leiter
LA
, McGuire
DK
, Wilding
JPH
, Ruff
CT
, Gause-Nilsson
IAM
, Fredriksson
M
, Johansson
PA
, Langkilde
A-M
, Sabatine
MS.
Dapagliflozin and cardiovascular outcomes in type 2 diabetes
.
N Engl J Med
2019
;
380
:
347
–
357
.
576Neal
B
, Perkovic
V
, Mahaffey
KW
, De Zeeuw
D
, Fulcher
G
, Erondu
N
, Shaw
W
, Law
G
, Desai
M
, Matthews
DR.
Canagliflozin and cardiovascular and renal events in type 2 diabetes
.
N Engl J Med
2017
;
377
:
644
–
657
.
577Adingupu
DD
, Göpel
SO
, Grönros
J
, Behrendt
M
, Sotak
M
, Miliotis
T
, Dahlqvist
U
, Gan
L-M
, Jönsson-Rylander
A-C.
SGLT2 inhibition with empagliflozin improves coronary microvascular function and cardiac contractility in prediabetic ob/ob(-/-) mice
.
Cardiovasc Diabetol
2019
;
18
:
16
.
578Lee
HC
, Shiou
YL
, Jhuo
SJ
, Chang
CY
, Liu
PL
, Jhuang
WJ
, Dai
ZK
, Chen
WY
, Chen
YF
, Lee
AS.
The sodium-glucose co-transporter 2 inhibitor empagliflozin attenuates cardiac fibrosis and improves ventricular hemodynamics in hypertensive heart failure rats
.
Cardiovasc Diabetol
2019
;
18
:
45
.
579Lee
TM
, Chang
NC
, Lin
SZ.
Dapagliflozin, a selective SGLT2 Inhibitor, attenuated cardiac fibrosis by regulating the macrophage polarization via STAT3 signaling in infarcted rat hearts
.
Free Radic Biol Med
2017
;
104
:
298
–
310
.
580Lim
VG
, Bell
RM
, Arjun
S
, Kolatsi-Joannou
M
, Long
DA
, Yellon
DM.
SGLT2 inhibitor, canagliflozin, attenuates myocardial infarction in the diabetic and nondiabetic heart
.
JACC Basic Transl Sci
2019
;
4
:
15
–
26
.
581Hippisley-Cox
J
, Coupland
C.
Diabetes treatments and risk of heart failure, cardiovascular disease, and all cause mortality: cohort study in primary care
.
BMJ
2016
;
354
:
i3477
.
582Investigators
OT
, Gerstein
HC
, Bosch
J
, Dagenais
GR
, Diaz
R
, Jung
H
, Maggioni
AP
, Pogue
J
, Probstfield
J
, Ramachandran
A
, Riddle
MC
, Ryden
LE
, Yusuf
S.
Basal insulin and cardiovascular and other outcomes in dysglycemia
.
N Engl J Med
2012
;
367
:
319
–
328
.
583Marso
SP
, McGuire
DK
, Zinman
B
, Poulter
NR
, Emerson
SS
, Pieber
TR
, Pratley
RE
, Haahr
PM
, Lange
M
, Brown-Frandsen
K
, Moses
A
, Skibsted
S
, Kvist
K
, Buse
JB
; DEVOTE Study Group.
Efficacy and safety of degludec versus glargine in type 2 diabetes
.
N Engl J Med
2017
;
377
:
723
–
732
.
584Shiuchi
T
, Cui
TX
, Wu
L
, Nakagami
H
, Takeda-Matsubara
Y
, Iwai
M
, Horiuchi
M.
ACE inhibitor improves insulin resistance in diabetic mouse via bradykinin and NO
.
Hypertension
2002
;
40
:
329
–
334
.
585Torlone
E
, Britta
M
, Rambotti
AM
, Perriello
G
, Santeusanio
F
, Brunetti
P
, Bolli
GB.
Improved insulin action and glycemic control after long-term angiotensin-converting enzyme inhibition in subjects with arterial hypertension and type II diabetes
.
Diabetes Care
1993
;
16
:
1347
–
1355
.
586Alkharouf
J
, Nalinikumari
K
, Corry
D
, Tuck
M.
Long-term effects of the angiotensin converting enzyme inhibitor captopril on metabolic control in non-insulin-dependent diabetes mellitus
.
Am J Hypertens
1993
;
6
:
337
–
343
.
587Morris
AD
, Boyle
DI
, McMahon
AD
, Pearce
H
, Evans
JM
, Newton
RW
, Jung
RT
, MacDonald
TM.
ACE inhibitor use is associated with hospitalization for severe hypoglycemia in patients with diabetes. DARTS/MEMO Collaboration. Diabetes Audit and Research in Tayside, Scotland. Medicines Monitoring Unit
.
Diabetes Care
1997
;
20
:
1363
–
1367
.
588Seshadri
S
, Rapaka
N
, Prajapati
B
, Mandaliya
D
, Patel
S
, Muggalla
CS
, Kapadia
B
, Babu
PP
, Misra
P
, Saxena
U.
Statins exacerbate glucose intolerance and hyperglycemia in a high sucrose fed rodent model
.
Sci Rep
2019
;
9
:
8825
.
589Erqou
S
, Lee
CC
, Adler
AI.
Statins and glycaemic control in individuals with diabetes: a systematic review and meta-analysis
.
Diabetologia
2014
;
57
:
2444
–
2452
.
590Cui
JY
, Zhou
RR
, Han
S
, Wang
TS
, Wang
LQ
, Xie
XH.
Statin therapy on glycemic control in type 2 diabetic patients: a network meta-analysis
.
J Clin Pharm Ther
2018
;
43
:
556
–
570
.
591Kim
J
, Lee
HS
, Lee
KY.
Effect of statins on fasting glucose in non-diabetic individuals: nationwide population-based health examination in Korea
.
Cardiovasc Diabetol
2018
;
17
:
155
.
592Struthers
AD
, Murphy
MB
, Dollery
CT.
Glucose tolerance during antihypertensive therapy in patients with diabetes mellitus
.
Hypertension
1985
;
7
:
II95
–
101
.
593Vue
MH
, Setter
SM.
Drug-induced glucose alterations part 1: drug-induced hypoglycemia
.
Diabetes Spectr
2011
;
24
:
171
–
177
.
594Dungan
K
, Merrill
J
, Long
C
, Binkley
P.
Effect of beta blocker use and type on hypoglycemia risk among hospitalized insulin requiring patients
.
Cardiovasc Diabetol
2019
;
18
:
163
.
595Wright
AD
, Barber
SG
, Kendall
MJ
, Poole
PH.
Beta-adrenoceptor-blocking drugs and blood sugar control in diabetes mellitus
.
Br Med J
1979
;
1
:
159
–
161
.
596Bakris
GL
, Fonseca
V
, Katholi
RE
, McGill
JB
, Messerli
FH
, Phillips
RA
, Raskin
P
, Wright
JT
Jr, Oakes
R
, Lukas
MA
, Anderson
KM
, Bell
DS
; GEMINI Investigators.
Metabolic effects of carvedilol vs metoprolol in patients with type 2 diabetes mellitus and hypertension: a randomized controlled trial
.
JAMA
2004
;
292
:
2227
–
2236
.
597Sirenko
Y.
Effect of beta-blockers on insulin resistance in patients with hypertension and metabolic syndrome after 6 months of treatment
.
J Endocrinol Diabetes
2017
;
4
:
1
–
11
.
598Izawa
H
, Murohara
T
, Nagata
K
, Isobe
S
, Asano
H
, Amano
T
, Ichihara
S
, Kato
T
, Ohshima
S
, Murase
Y
, Iino
S
, Obata
K
, Noda
A
, Okumura
K
, Yokota
M.
Mineralocorticoid receptor antagonism ameliorates left ventricular diastolic dysfunction and myocardial fibrosis in mildly symptomatic patients with idiopathic dilated cardiomyopathy: a pilot study
.
Circulation
2005
;
112
:
2940
–
2945
.
599Mottram
PM
, Haluska
B
, Leano
R
, Cowley
D
, Stowasser
M
, Marwick
TH.
Effect of aldosterone antagonism on myocardial dysfunction in hypertensive patients with diastolic heart failure
.
Circulation
2004
;
110
:
558
–
565
.
600Kosmala
W
, Przewlocka-Kosmala
M
, Szczepanik-Osadnik
H
, Mysiak
A
, O'Moore-Sullivan
T
, Marwick
TH.
A randomized study of the beneficial effects of aldosterone antagonism on LV function, structure, and fibrosis markers in metabolic syndrome
.
JACC Cardiovasc Imaging
2011
;
4
:
1239
–
1249
.
601Srinivasa
S
, Fitch
KV
, Wong
K
, O'Malley
TK
, Maehler
P
, Branch
KL
, Looby
SE
, Burdo
TH
, Martinez-Salazar
EL
, Torriani
M
, Lyons
SH
, Weiss
J
, Feldpausch
M
, Stanley
TL
, Adler
GK
, Grinspoon
SK.
Randomized, placebo-controlled trial to evaluate effects of eplerenone on metabolic and inflammatory indices in HIV
.
J Clin Endocrinol Metab
2018
;
103
:
2376
–
2384
.
602Nagatomo
Y
, Meguro
T
, Ito
H
, Koide
K
, Anzai
T
, Fukuda
K
, Ogawa
S
, Yoshikawa
T.
Significance of AT1 receptor independent activation of mineralocorticoid receptor in murine diabetic cardiomyopathy
.
PLoS One
2014
;
9
:
e93145
.
603Ramírez
E
, Klett-Mingo
M
, Ares-Carrasco
S
, Picatoste
B
, Ferrarini
A
, Rupérez
FJ
, Caro-Vadillo
A
, Barbas
C
, Egido
J
, Tuñón
J
, Lorenzo
Ó.
Eplerenone attenuated cardiac steatosis, apoptosis and diastolic dysfunction in experimental type-II diabetes
.
Cardiovasc Diabetol
2013
;
12
:
172
.
604Liu
W
, Gong
W
, He
M
, Liu
Y
, Yang
Y
, Wang
M
, Wu
M
, Guo
S
, Yu
Y
, Wang
X
, Sun
F
, Li
Y
, Zhou
L
, Qin
S
, Zhang
Z.
Spironolactone protects against diabetic cardiomyopathy in streptozotocin-induced diabetic rats
.
J Diabetes Res
2018
;
2018
:
9232065
.
605Zhang
M
, Gu
H
, Xu
W
, Zhou
X.
Down-regulation of lncRNA MALAT1 reduces cardiomyocyte apoptosis and improves left ventricular function in diabetic rats
.
Int J Cardiol
2016
;
203
:
214
–
216
.
606Zhao
F
, Li
B
, Wei
YZ
, Zhou
B
, Wang
H
, Chen
M
, Gan
XD
, Wang
ZH
, Xiong
SX.
MicroRNA-34a regulates high glucose-induced apoptosis in H9c2 cardiomyocytes
.
J Huazhong Univ Sci Technol Med Sci
2013
;
33
:
834
–
839
.
607Zheng
D
, Zhang
Y
, Hu
Y
, Guan
J
, Xu
L
, Xiao
W
, Zhong
Q
, Ren
C
, Lu
J
, Liang
J
, Hou
J.
Long noncoding RNA Crnde attenuates cardiac fibrosis via Smad3-Crnde negative feedback in diabetic cardiomyopathy
.
FEBS J
2019
;
286
:
1645
–
1655
.
608Li
X
, Du
N
, Zhang
Q
, Li
J
, Chen
X
, Liu
X
, Hu
Y
, Qin
W
, Shen
N
, Xu
C
, Fang
Z
, Wei
Y
, Wang
R
, Du
Z
, Zhang
Y
, Lu
Y.
MicroRNA-30d regulates cardiomyocyte pyroptosis by directly targeting foxo3a in diabetic cardiomyopathy
.
Cell Death Dis
2014
;
5
:
e1479
.
609Yi
F
, Shang
YG
, Li
B
, Dai
SL
, Wu
W
, Cheng
L
, Wang
XC.
MicroRNA-193-5p modulates angiogenesis through IGF2 in type 2 diabetic cardiomyopathy
.
Biochem Biophys Res Commun
2017
;
491
:
876
–
882
.
610Chen
C
, Yang
SL
, Li
HP
, Yin
ZW
, Fan
JH
, Zhao
YR
, Gong
W
, Yan
MW
, Wang
DW.
Mir30c is involved in diabetic cardiomyopathy through regulation of cardiac autophagy via BECN1
.
Mol Ther Nucleic Acids
2017
;
7
:
127
–
139
.
611Yang
Z
, Wang
M
, Zhang
Y
, Cai
F
, Jiang
B
, Zha
W
, Yu
W.
Metformin ameliorates diabetic cardiomyopathy by activating the PK2/PKR pathway
.
Front Physiol
2020
;
11
:
425
.
612Halabi
A
, Sen
J
, Huynh
Q
, Marwick
TH.
Metformin treatment in heart failure with preserved ejection fraction: a systematic review and meta-regression analysis
.
Cardiovasc Diabetol
2020
;
19
:
124
.
613Bergmark
BA
, Bhatt
DL
, McGuire
DK
, Cahn
A
, Mosenzon
O
, Steg
PG
, Im
K
, Kanevsky
E
, Gurmu
Y
, Raz
I
, Braunwald
E
, Scirica
BM
; SAVOR-TIMI 53 Steering Committee and Investigators.
Metformin use and clinical outcomes among patients with diabetes mellitus with or without heart failure or kidney dysfunction observations from the SAVOR-TIMI 53 Trial
.
Circ
2019
;
140
:
1004
–
1014
.
614Bromage
DI
, Godec
TR
, Pujades-Rodriguez
M
, Gonzalez-Izquierdo
A
, Denaxas
S
, Hemingway
H
, Yellon
DM.
Metformin use and cardiovascular outcomes after acute myocardial infarction in patients with type 2 diabetes: a cohort study
.
Cardiovasc Diabetol
2019
;
18
:
019
–
0972
.
615Ye
YM
, Keyes
KT
, Zhang
CF
, Perez-Polo
JR
, Lin
Y
, Birnbaum
Y.
The myocardial infarct size-limiting effect of sitagliptin is PKA-dependent, whereas the protective effect of pioglitazone is partially dependent on PKA
.
Am J Physiol Heart Circ Physiol
2010
;
298
:
H1454
–
H1465
.
616Liu
Q
, Anderson
C
, Broyde
A
, Polizzi
C
, Fernandez
R
, Baron
A
, Parkes
DG.
Glucagon-like peptide-1 and the exenatide analogue AC3174 improve cardiac function, cardiac remodeling, and survival in rats with chronic heart failure
.
Cardiovasc Diabetol
2010
;
9
:
76
.
617Sonne
DP
, Engstrom
T
, Treiman
M.
Protective effects of GLP-1 analogues exendin-4 and GLP-1(9-36) amide against ischemia-reperfusion injury in rat heart
.
Regul Pept
2008
;
146
:
243
–
249
.
618Noyan-Ashraf
MH
, Momen
MA
, Ban
K
, Sadi
AM
, Zhou
YQ
, Riazi
AM
, Baggio
LL
, Henkelman
RM
, Husain
M
, Drucker
DJ.
GLP-1R agonist liraglutide activates cytoprotective pathways and improves outcomes after experimental myocardial infarction in mice
.
Diabetes
2009
;
58
:
975
–
983
.
619Kim
JA
, Wei
Y
, Sowers
JR.
Role of mitochondrial dysfunction in insulin resistance
.
Circ Res
2008
;
102
:
401
–
414
.
620Zhao
JV
, Xu
L
, Lin
SL
, Schooling
CM.
Spironolactone and glucose metabolism, a systematic review and meta-analysis of randomized controlled trials
.
J Am Soc Hypertens
2016
;
10
:
671
–
682
.
621Yamaji
M
, Tsutamoto
T
, Kawahara
C
, Nishiyama
K
, Yamamoto
T
, Fujii
M
, Horie
M.
Effect of eplerenone versus spironolactone on cortisol and hemoglobin A(1)(c) levels in patients with chronic heart failure
.
Am Heart J
2010
;
160
:
915
–
921
.
Published on behalf of the European Society of Cardiology. All rights reserved. © The Author(s) 2021. For permissions, please email:
[email protected].