Abstract

Diabetic cardiomyopathy (CM), occurring in the absence of hypertension, coronary artery disease, and valvular or congenital heart disease, is now recognized as a distinct, multifactorial disease leading to ventricular hypertrophy and abnormal myocardial contractility that correlates with an array of complex molecular and cellular changes. Animal models provide the unique opportunity to investigate mechanistic aspects of diabetic CM, but important caveats exist when extrapolating findings obtained from preclinical models of diabetes to humans. Indeed, animal models do not recapitulate the complexity of environmental factors, most notably the duration of the exposure to insulin resistance that may play a crucial role in the development of diabetic CM. Moreover, most preclinical studies are performed in animals with uncontrolled or poorly controlled diabetes, whereas patients tend to undergo therapeutic intervention. Finally, whilst type 2 diabetes mellitus prevalence trajectory mainly increases at 40- < 75 years (with a currently alarming increase at younger ages, however), it is a legitimate concern how closely rodent models employing young animals recapitulate the disease developing in old people. The aim of this review is to identify the current limitations of rodent models and to discuss how future mechanistic and preclinical studies should integrate key confounding factors to better mimic the diabetic CM phenotype.

1. Introduction

The prevalence of type 2 diabetes mellitus (T2DM) is increasing worldwide, afflicting all ages, sexes, and socioeconomic classes, ultimately leading to frailty and unhealthy ageing.1 Cardiovascular (CV) complications are the leading causes of morbidity and mortality in T2DM patients, accounting for about two-thirds of overall deaths as evidenced by the Framingham Heart Study.2 One of the specific CV complications in T2DM is diabetic cardiomyopathy (CM), originally described as an early diastolic dysfunction progressing to systolic dysfunction and heart failure (HF) in the absence of hypertension, coronary artery disease, and valvular or congenital heart disease.3 The CARDIA study provided strong evidence for the diagnosis of diabetic CM,4 identifying subtle diastolic and/or systolic myocardial abnormalities preceding the onset of overt CM and HF.5–7

Diabetic CM is now recognized as a distinct, multifactorial disease leading to ventricular hypertrophy and abnormal myocardial contractility that correlates with an array of complex molecular and cellular changes (Figure 1).8

Mechanisms contributing to cardiomyocyte dysfunction in diabetic cardiomyopathy. Hormonal and metabolic alterations may result in hyperglycaemia, insulin resistance, and lipid overload which cause through specific signalling pathways subcellular component abnormalities. This includes mitochondrial dysfunction, impaired metabolic flexibility, Ca2 + dysregulation, and activation of gene transcription programme involved in cardiac remodelling and senescence. These molecular and cellular events contribute to diastolic and systolic dysfunction. GPCR, G protein-coupled receptor; IR, insulin receptor; ROS, reactive oxygen species; RyR, ryanodine receptor.
Figure 1

Mechanisms contributing to cardiomyocyte dysfunction in diabetic cardiomyopathy. Hormonal and metabolic alterations may result in hyperglycaemia, insulin resistance, and lipid overload which cause through specific signalling pathways subcellular component abnormalities. This includes mitochondrial dysfunction, impaired metabolic flexibility, Ca2 + dysregulation, and activation of gene transcription programme involved in cardiac remodelling and senescence. These molecular and cellular events contribute to diastolic and systolic dysfunction. GPCR, G protein-coupled receptor; IR, insulin receptor; ROS, reactive oxygen species; RyR, ryanodine receptor.

With no pathognomonic feature identified as specific to human diabetic CM, however, asserting the ‘uniqueness’ of diabetic CM will require a thorough consideration for the «metabolic exposome», including diet, lifestyle, glycaemia, obesity, sedentary behaviour, alongside unmodifiable confounders, such as genetic susceptibility, sex, and ageing, which are intertwined in the pathogenesis of diabetic CM.9

The complex impact of these factors are then magnified by other processes linking the heart to the functional state of key metabolic organs, i.e. the adipose tissue, liver, kidney, and the gut (microbiota) that may separately exert noxious cardiac effects through a crosstalk mediated by e.g. proinflammatory cytokines, profibrotic factors, microvesicles, miRNAs, and immune cells. This endocrine organ-crosstalk evolves into a paracrine cellular-crosstalk between cardiomyocytes, fibroblasts, endothelial cells, and immune cells in the myocardium (Figure 2).

Diabetic heart is at the cross-road of environmental factors, organ-crosstalk, and paracrine cellular-crosstalk between cardiomyocytes, fibroblasts, endothelial cells, and immune cells in the myocardium.
Figure 2

Diabetic heart is at the cross-road of environmental factors, organ-crosstalk, and paracrine cellular-crosstalk between cardiomyocytes, fibroblasts, endothelial cells, and immune cells in the myocardium.

Whilst animal models provide the unique opportunity to investigate mechanistic aspects of diabetic complications, including diabetic CM, important caveats exist when extrapolating findings obtained from preclinical models of diabetes to humans because animal models do not recapitulate the full complexity of diabetic CM. Common features, such as insulin, glucose levels, and dyslipidaemia occurring in diabetic CM, are generally well reproduced in most rodent models of diabetes (Table 1), but there are many open questions with clinical significance. For example, there is a clear lack of molecular data in human diabetic CM, to which we could compare findings obtained in rodent models. This is a great obstacle, as therapeutic responsiveness of mice and humans with diabetic CM diverge. Specifically, strict glycaemic control protects rodents from HF,63,64 but not humans,65 some antidiabetic agents even increase the risk for HF.66–69 Moreover, we do not have any models to predict which diabetic patient will develop diabetic CM, what the causal factors to promote either HF with reduced ejection fraction (HFrEF) or HF with preserved ejection fraction (HFpEF) from diabetic CM are, and to understand why strict glycaemic control does not ameliorate diabetic CM, etc. Failure to answer these burning questions suggest the possibility that there are important pathogenic stimuli in human patients with diabetic CM that are poorly reproduced by current rodent models.70 Obvious additional species-specific differences include but do not limit to chronicity of insulin resistance, differences in cardiac physiology, such as heart rate, Ca2+ fluxes, sarcomere composition and vessel function, resistance to developing micro- and macrovascular diseases in rodents, differential hormonal milieu, concentrations of various lipid species,71 and control of diabetes (typically no in preclinical models vs. patients). Furthermore, whilst T2DM is traditionally a disease of the elderly in humans (i.e. 40 to < 75 years with increasing prevalence in the younger population,72,73 however), most rodent models employ young adult animals. Importantly, diabetic CM emerges in the midst of multiple organ disorders, that may significantly alter cardiac function by modest yet chronic changes in ion concentration, pH, circulating abnormal proteins and metabolites, subclinical increase in afterload, liver dysfunction, skeletal muscle dysfunction, presence of obstructive sleep apnoea (OSA), etc. However, current rodent models poorly reproduce such common comorbid conditions.

Table 1

Rodent models that recapitulate diabetic cardiomyopathy features found in humans

Animal modelCardiac/noncardiac alterations that recapitulate human features of diabetic cardiomyopathyReferences
MiceHFDC57BL6 miceEarly onset of metabolic alterations and cardiac LV dysfunction (5 weeks after starting HFD), obesity, hyperglycaemia, hyperinsulinemia, dyslipidaemia. Fatty liver, combined visceral/subcutaneous adiposity with increased rate of crown-like structures, mild diabetic nephropathy.10–14
C57BL6/J mice + HFD + angiotensin II infusion.Model of HFpEF (LV hypertrophy and LV diastolic dysfunction; no change in LVEF).12
ob/ob miceLV diastolic dysfunction and features of lipotoxicity. Reduced circulating leptin, excessive food intake, increased insulin, hyperglycaemia, hyperinsulinemia, and triglyceride levels. Compromised immune system, reproductive ability, altered incidence of malignancies (↑ or ↓).15–17
C57BL/6N mice + HFD + po. L-NAME administrationModel of HFpEF (hypertrophic response, diastolic dysfunction, pulmonary congestion, reduction in contraction velocity and impaired relaxation). Reduced skeletal muscle strength.18
Models of lipotoxicityAlteration in myosin heavy chain acyl-CoA synthetase (MHC-ACS mice)Lipotoxicity, myocardial macrophage infiltration, inflammation, abnormal cardiac metabolism, cardiac hypertrophy, LV dysfunction and premature death.19,20
GPI-anchored human lipoprotein lipase transgenic mice (hLpLGPI mice)Lipotoxicity, cardiac hypertrophy, abnormal cardiac metabolism, LV dysfunction, and cardiac fibrosis.21
Myosin heavy chain-peroxisome proliferator-activated receptor α mice (MHC-PPARα mice)Lipotoxicity, cardiac hypertrophy, abnormal cardiac metabolism, LV dysfunction, and cardiac fibrosis.22
Myosin heavy chain fatty acid transport protein mice (MHC-FATP mice)Lipotoxicity, LV diastolic dysfunction and prolonged QTc intervals.23,24
Adipose TG lipase knockout miceLipotoxicity. LV dysfunction and premature death. Reduced triglyceride hydrolase activity in skeletal muscle and adipose tissue. Reduced glycogen content in liver.25
T1DMStreptozotocin Intraperitoneal routeReduction in heart rate, amplitude of contraction and of ventricular pressure, and prolongation on the rate of ventricular myocyte contraction and relaxation. Kidney enlargement. Reduced body weight and circulating insulin levels.26–31
Intravenous route32
T2DMdb/db miceDecreased systolic function, abnormal diastolic filling, and electrophysiological alterations. Leptin receptor deficiency due to a point mutation. Hyperphagia, dyslipidaemia, progressive diabetic nephropathy.33–35
RatHFDSprague-Dawley ratsLipotoxicity, cardiac fibrosis and hypertrophy. Increased plasma triglyceride, cholesterol and LDL, reduced HDL levels. Increased circulating markers of oxidative stress and inflammation.36,37
ObesityObese Zucker rats (fa/fa)Lipotoxicity and increased LV end-diastolic volume and stroke volume. Reduced cardiac levels of taurine, glutamate, glutamine, and glutathione; increased cardiac lactate levels. Primarily subcutaneous obesity.38,39
DahlS.Z-Lepr(fa)/Lepr(fa) (DS/obese) ratsLV diastolic dysfunction, LV hypertrophy, and cardiac fibrosis, oxidative stress, and inflammation. Increased body weight, subcutaneous and visceral fat mass. Elevated serum insulin, LDL/HDL ratio and triglyceride levels.40
T1DMStreptozotocin- Intraperitoneal routeLV systolic and diastolic dysfunction, oxidative stress increased rate of apoptosis, mitochondrial damage, and fibrosis. Reduced body weight, increased circulating glucose and HbA1c levels.41–44
- Intravenous routeReduced LV systolic and diastolic function. Polydipsia, polyuria, glycosuria, proteinuria, uraemia.45,46
T2DMZucker diabetic fatty rats (ZDF)Increased heart and LV weight, presence of fibrosis, depressed RV and LV systolic function. Dyslipidaemia. Respiratory muscle weakness. Diabetic neuropathy, microangiopathy, nephropathy, hypercoagulability.47–53
Goto-Kakizaki rats (GK) Intraperitoneal injection of Streptozotocin + nicotinamideCardiac hypertrophy, increased extracellular matrix deposition and increased heart size. Non-obese model of T2DM with moderate hepatic triglyceride accumulation. Age-dependent development of glomerulosclerosis. Hepatic lipotoxicity (increased accumulation of triglycerides, cholesterol, and free fatty acids). Increased serum and hepatic lipid peroxidation.54–60
Otsuka–Long–Evans–Tokushima fatty (OLETF) ratsLate-onset hyperglycaemia, mild obesity, diabetes mostly in males, multiple recessive genes involved, age-dependent atrophy of pancreatic islets, diabetic nephropathy, primarily visceral obesity.39,61,62
Animal modelCardiac/noncardiac alterations that recapitulate human features of diabetic cardiomyopathyReferences
MiceHFDC57BL6 miceEarly onset of metabolic alterations and cardiac LV dysfunction (5 weeks after starting HFD), obesity, hyperglycaemia, hyperinsulinemia, dyslipidaemia. Fatty liver, combined visceral/subcutaneous adiposity with increased rate of crown-like structures, mild diabetic nephropathy.10–14
C57BL6/J mice + HFD + angiotensin II infusion.Model of HFpEF (LV hypertrophy and LV diastolic dysfunction; no change in LVEF).12
ob/ob miceLV diastolic dysfunction and features of lipotoxicity. Reduced circulating leptin, excessive food intake, increased insulin, hyperglycaemia, hyperinsulinemia, and triglyceride levels. Compromised immune system, reproductive ability, altered incidence of malignancies (↑ or ↓).15–17
C57BL/6N mice + HFD + po. L-NAME administrationModel of HFpEF (hypertrophic response, diastolic dysfunction, pulmonary congestion, reduction in contraction velocity and impaired relaxation). Reduced skeletal muscle strength.18
Models of lipotoxicityAlteration in myosin heavy chain acyl-CoA synthetase (MHC-ACS mice)Lipotoxicity, myocardial macrophage infiltration, inflammation, abnormal cardiac metabolism, cardiac hypertrophy, LV dysfunction and premature death.19,20
GPI-anchored human lipoprotein lipase transgenic mice (hLpLGPI mice)Lipotoxicity, cardiac hypertrophy, abnormal cardiac metabolism, LV dysfunction, and cardiac fibrosis.21
Myosin heavy chain-peroxisome proliferator-activated receptor α mice (MHC-PPARα mice)Lipotoxicity, cardiac hypertrophy, abnormal cardiac metabolism, LV dysfunction, and cardiac fibrosis.22
Myosin heavy chain fatty acid transport protein mice (MHC-FATP mice)Lipotoxicity, LV diastolic dysfunction and prolonged QTc intervals.23,24
Adipose TG lipase knockout miceLipotoxicity. LV dysfunction and premature death. Reduced triglyceride hydrolase activity in skeletal muscle and adipose tissue. Reduced glycogen content in liver.25
T1DMStreptozotocin Intraperitoneal routeReduction in heart rate, amplitude of contraction and of ventricular pressure, and prolongation on the rate of ventricular myocyte contraction and relaxation. Kidney enlargement. Reduced body weight and circulating insulin levels.26–31
Intravenous route32
T2DMdb/db miceDecreased systolic function, abnormal diastolic filling, and electrophysiological alterations. Leptin receptor deficiency due to a point mutation. Hyperphagia, dyslipidaemia, progressive diabetic nephropathy.33–35
RatHFDSprague-Dawley ratsLipotoxicity, cardiac fibrosis and hypertrophy. Increased plasma triglyceride, cholesterol and LDL, reduced HDL levels. Increased circulating markers of oxidative stress and inflammation.36,37
ObesityObese Zucker rats (fa/fa)Lipotoxicity and increased LV end-diastolic volume and stroke volume. Reduced cardiac levels of taurine, glutamate, glutamine, and glutathione; increased cardiac lactate levels. Primarily subcutaneous obesity.38,39
DahlS.Z-Lepr(fa)/Lepr(fa) (DS/obese) ratsLV diastolic dysfunction, LV hypertrophy, and cardiac fibrosis, oxidative stress, and inflammation. Increased body weight, subcutaneous and visceral fat mass. Elevated serum insulin, LDL/HDL ratio and triglyceride levels.40
T1DMStreptozotocin- Intraperitoneal routeLV systolic and diastolic dysfunction, oxidative stress increased rate of apoptosis, mitochondrial damage, and fibrosis. Reduced body weight, increased circulating glucose and HbA1c levels.41–44
- Intravenous routeReduced LV systolic and diastolic function. Polydipsia, polyuria, glycosuria, proteinuria, uraemia.45,46
T2DMZucker diabetic fatty rats (ZDF)Increased heart and LV weight, presence of fibrosis, depressed RV and LV systolic function. Dyslipidaemia. Respiratory muscle weakness. Diabetic neuropathy, microangiopathy, nephropathy, hypercoagulability.47–53
Goto-Kakizaki rats (GK) Intraperitoneal injection of Streptozotocin + nicotinamideCardiac hypertrophy, increased extracellular matrix deposition and increased heart size. Non-obese model of T2DM with moderate hepatic triglyceride accumulation. Age-dependent development of glomerulosclerosis. Hepatic lipotoxicity (increased accumulation of triglycerides, cholesterol, and free fatty acids). Increased serum and hepatic lipid peroxidation.54–60
Otsuka–Long–Evans–Tokushima fatty (OLETF) ratsLate-onset hyperglycaemia, mild obesity, diabetes mostly in males, multiple recessive genes involved, age-dependent atrophy of pancreatic islets, diabetic nephropathy, primarily visceral obesity.39,61,62

HFD, high-fat diet; LV, left ventricle; LVEF, left ventricular ejection fraction; T1DM, type 1 diabetes mellitus; T2DM, type 2 diabetes mellitus.

Table 1

Rodent models that recapitulate diabetic cardiomyopathy features found in humans

Animal modelCardiac/noncardiac alterations that recapitulate human features of diabetic cardiomyopathyReferences
MiceHFDC57BL6 miceEarly onset of metabolic alterations and cardiac LV dysfunction (5 weeks after starting HFD), obesity, hyperglycaemia, hyperinsulinemia, dyslipidaemia. Fatty liver, combined visceral/subcutaneous adiposity with increased rate of crown-like structures, mild diabetic nephropathy.10–14
C57BL6/J mice + HFD + angiotensin II infusion.Model of HFpEF (LV hypertrophy and LV diastolic dysfunction; no change in LVEF).12
ob/ob miceLV diastolic dysfunction and features of lipotoxicity. Reduced circulating leptin, excessive food intake, increased insulin, hyperglycaemia, hyperinsulinemia, and triglyceride levels. Compromised immune system, reproductive ability, altered incidence of malignancies (↑ or ↓).15–17
C57BL/6N mice + HFD + po. L-NAME administrationModel of HFpEF (hypertrophic response, diastolic dysfunction, pulmonary congestion, reduction in contraction velocity and impaired relaxation). Reduced skeletal muscle strength.18
Models of lipotoxicityAlteration in myosin heavy chain acyl-CoA synthetase (MHC-ACS mice)Lipotoxicity, myocardial macrophage infiltration, inflammation, abnormal cardiac metabolism, cardiac hypertrophy, LV dysfunction and premature death.19,20
GPI-anchored human lipoprotein lipase transgenic mice (hLpLGPI mice)Lipotoxicity, cardiac hypertrophy, abnormal cardiac metabolism, LV dysfunction, and cardiac fibrosis.21
Myosin heavy chain-peroxisome proliferator-activated receptor α mice (MHC-PPARα mice)Lipotoxicity, cardiac hypertrophy, abnormal cardiac metabolism, LV dysfunction, and cardiac fibrosis.22
Myosin heavy chain fatty acid transport protein mice (MHC-FATP mice)Lipotoxicity, LV diastolic dysfunction and prolonged QTc intervals.23,24
Adipose TG lipase knockout miceLipotoxicity. LV dysfunction and premature death. Reduced triglyceride hydrolase activity in skeletal muscle and adipose tissue. Reduced glycogen content in liver.25
T1DMStreptozotocin Intraperitoneal routeReduction in heart rate, amplitude of contraction and of ventricular pressure, and prolongation on the rate of ventricular myocyte contraction and relaxation. Kidney enlargement. Reduced body weight and circulating insulin levels.26–31
Intravenous route32
T2DMdb/db miceDecreased systolic function, abnormal diastolic filling, and electrophysiological alterations. Leptin receptor deficiency due to a point mutation. Hyperphagia, dyslipidaemia, progressive diabetic nephropathy.33–35
RatHFDSprague-Dawley ratsLipotoxicity, cardiac fibrosis and hypertrophy. Increased plasma triglyceride, cholesterol and LDL, reduced HDL levels. Increased circulating markers of oxidative stress and inflammation.36,37
ObesityObese Zucker rats (fa/fa)Lipotoxicity and increased LV end-diastolic volume and stroke volume. Reduced cardiac levels of taurine, glutamate, glutamine, and glutathione; increased cardiac lactate levels. Primarily subcutaneous obesity.38,39
DahlS.Z-Lepr(fa)/Lepr(fa) (DS/obese) ratsLV diastolic dysfunction, LV hypertrophy, and cardiac fibrosis, oxidative stress, and inflammation. Increased body weight, subcutaneous and visceral fat mass. Elevated serum insulin, LDL/HDL ratio and triglyceride levels.40
T1DMStreptozotocin- Intraperitoneal routeLV systolic and diastolic dysfunction, oxidative stress increased rate of apoptosis, mitochondrial damage, and fibrosis. Reduced body weight, increased circulating glucose and HbA1c levels.41–44
- Intravenous routeReduced LV systolic and diastolic function. Polydipsia, polyuria, glycosuria, proteinuria, uraemia.45,46
T2DMZucker diabetic fatty rats (ZDF)Increased heart and LV weight, presence of fibrosis, depressed RV and LV systolic function. Dyslipidaemia. Respiratory muscle weakness. Diabetic neuropathy, microangiopathy, nephropathy, hypercoagulability.47–53
Goto-Kakizaki rats (GK) Intraperitoneal injection of Streptozotocin + nicotinamideCardiac hypertrophy, increased extracellular matrix deposition and increased heart size. Non-obese model of T2DM with moderate hepatic triglyceride accumulation. Age-dependent development of glomerulosclerosis. Hepatic lipotoxicity (increased accumulation of triglycerides, cholesterol, and free fatty acids). Increased serum and hepatic lipid peroxidation.54–60
Otsuka–Long–Evans–Tokushima fatty (OLETF) ratsLate-onset hyperglycaemia, mild obesity, diabetes mostly in males, multiple recessive genes involved, age-dependent atrophy of pancreatic islets, diabetic nephropathy, primarily visceral obesity.39,61,62
Animal modelCardiac/noncardiac alterations that recapitulate human features of diabetic cardiomyopathyReferences
MiceHFDC57BL6 miceEarly onset of metabolic alterations and cardiac LV dysfunction (5 weeks after starting HFD), obesity, hyperglycaemia, hyperinsulinemia, dyslipidaemia. Fatty liver, combined visceral/subcutaneous adiposity with increased rate of crown-like structures, mild diabetic nephropathy.10–14
C57BL6/J mice + HFD + angiotensin II infusion.Model of HFpEF (LV hypertrophy and LV diastolic dysfunction; no change in LVEF).12
ob/ob miceLV diastolic dysfunction and features of lipotoxicity. Reduced circulating leptin, excessive food intake, increased insulin, hyperglycaemia, hyperinsulinemia, and triglyceride levels. Compromised immune system, reproductive ability, altered incidence of malignancies (↑ or ↓).15–17
C57BL/6N mice + HFD + po. L-NAME administrationModel of HFpEF (hypertrophic response, diastolic dysfunction, pulmonary congestion, reduction in contraction velocity and impaired relaxation). Reduced skeletal muscle strength.18
Models of lipotoxicityAlteration in myosin heavy chain acyl-CoA synthetase (MHC-ACS mice)Lipotoxicity, myocardial macrophage infiltration, inflammation, abnormal cardiac metabolism, cardiac hypertrophy, LV dysfunction and premature death.19,20
GPI-anchored human lipoprotein lipase transgenic mice (hLpLGPI mice)Lipotoxicity, cardiac hypertrophy, abnormal cardiac metabolism, LV dysfunction, and cardiac fibrosis.21
Myosin heavy chain-peroxisome proliferator-activated receptor α mice (MHC-PPARα mice)Lipotoxicity, cardiac hypertrophy, abnormal cardiac metabolism, LV dysfunction, and cardiac fibrosis.22
Myosin heavy chain fatty acid transport protein mice (MHC-FATP mice)Lipotoxicity, LV diastolic dysfunction and prolonged QTc intervals.23,24
Adipose TG lipase knockout miceLipotoxicity. LV dysfunction and premature death. Reduced triglyceride hydrolase activity in skeletal muscle and adipose tissue. Reduced glycogen content in liver.25
T1DMStreptozotocin Intraperitoneal routeReduction in heart rate, amplitude of contraction and of ventricular pressure, and prolongation on the rate of ventricular myocyte contraction and relaxation. Kidney enlargement. Reduced body weight and circulating insulin levels.26–31
Intravenous route32
T2DMdb/db miceDecreased systolic function, abnormal diastolic filling, and electrophysiological alterations. Leptin receptor deficiency due to a point mutation. Hyperphagia, dyslipidaemia, progressive diabetic nephropathy.33–35
RatHFDSprague-Dawley ratsLipotoxicity, cardiac fibrosis and hypertrophy. Increased plasma triglyceride, cholesterol and LDL, reduced HDL levels. Increased circulating markers of oxidative stress and inflammation.36,37
ObesityObese Zucker rats (fa/fa)Lipotoxicity and increased LV end-diastolic volume and stroke volume. Reduced cardiac levels of taurine, glutamate, glutamine, and glutathione; increased cardiac lactate levels. Primarily subcutaneous obesity.38,39
DahlS.Z-Lepr(fa)/Lepr(fa) (DS/obese) ratsLV diastolic dysfunction, LV hypertrophy, and cardiac fibrosis, oxidative stress, and inflammation. Increased body weight, subcutaneous and visceral fat mass. Elevated serum insulin, LDL/HDL ratio and triglyceride levels.40
T1DMStreptozotocin- Intraperitoneal routeLV systolic and diastolic dysfunction, oxidative stress increased rate of apoptosis, mitochondrial damage, and fibrosis. Reduced body weight, increased circulating glucose and HbA1c levels.41–44
- Intravenous routeReduced LV systolic and diastolic function. Polydipsia, polyuria, glycosuria, proteinuria, uraemia.45,46
T2DMZucker diabetic fatty rats (ZDF)Increased heart and LV weight, presence of fibrosis, depressed RV and LV systolic function. Dyslipidaemia. Respiratory muscle weakness. Diabetic neuropathy, microangiopathy, nephropathy, hypercoagulability.47–53
Goto-Kakizaki rats (GK) Intraperitoneal injection of Streptozotocin + nicotinamideCardiac hypertrophy, increased extracellular matrix deposition and increased heart size. Non-obese model of T2DM with moderate hepatic triglyceride accumulation. Age-dependent development of glomerulosclerosis. Hepatic lipotoxicity (increased accumulation of triglycerides, cholesterol, and free fatty acids). Increased serum and hepatic lipid peroxidation.54–60
Otsuka–Long–Evans–Tokushima fatty (OLETF) ratsLate-onset hyperglycaemia, mild obesity, diabetes mostly in males, multiple recessive genes involved, age-dependent atrophy of pancreatic islets, diabetic nephropathy, primarily visceral obesity.39,61,62

HFD, high-fat diet; LV, left ventricle; LVEF, left ventricular ejection fraction; T1DM, type 1 diabetes mellitus; T2DM, type 2 diabetes mellitus.

The aim of this review is to identify the current limitations of rodent models and to discuss how future preclinical studies should integrate key confounding factors to better mimic the diabetic CM phenotype as it presents itself in clinic.

2. Molecular aspects of diabetic CM

Abnormalities responsible for hallmarks of diabetic CM, i.e. cardiac stiffness, hypertrophy, fibrosis, and ischaemia, eventually leading to HFpEF and/or HFrEF are highly complex. Both insulin resistance and chronic hyperglycaemia contribute to impaired cardiac contractility and structure via e.g. dysregulated intracellular Ca2+ homeostasis, abnormal PI3K/Akt pathway signalling, enhanced production of reactive oxygen species (ROS), advanced glycation end products (AGEs), cardiac protein O-GlcNAcylation, toxic fatty acid (FA) metabolites, as well as probably less well studied mechanisms, such as altered autophagy, and epigenetic dysregulation.74 These abnormalities do not emerge in isolation, but are interconnected. For example, the metabolic inflexibility in cardiomyocyte metabolism with a shift towards FA oxidation and ensuing mitochondrial ROS production can trigger endoplasmic reticulum (ER) stress, cardiomyocyte death, inflammation, and microvascular dysfunction.75

ER stress and mitochondrial dysfunction are key factors for the development and progression of diabetic CM. Altered Ca2+ handling is widely believed to underlie depressed contractility, slow relaxation, and arrhythmias triggered in diabetic CM.76 In murine models with diabetic CM, prolongation of intracellular Ca2+ decay and consequential decrease in Ca2+ transient amplitude directly correspond to delayed relaxation and abnormal contractility, respectively.76 The development of dysregulated Ca2+ cycling is facilitated by altered expression and/or activity of the L-type Ca2+ channels, ryanodine receptor, sarcoplasmic reticulum Ca2+ ATPase (SERCA2a), and Na+/Ca2 + exchanger (NCX). In T2DM models, these alterations hamper mitochondrial Ca2+ uptake, provoking an energy supply-and-demand mismatch with excessive mitochondrial ROS production.76 Moreover, ER stress, triggered by hyperglycaemia, free FAs, and inflammation, is an early event in diabetic CM, which may promote cardiomyocyte apoptosis and loss of function.77 Indeed, beyond changes in Ca2+-handling proteins per se, intercompartmental transfer of Ca2+ also occurs through the mitochondrial-associated membranes (MAMs; functional interaction sites between ER and mitochondria), exchanging lipids and Ca2+. In a diet-induced mouse model of diabetic CM recent evidence suggests that reticular-mitochondrial Ca2+ uncoupling is an early trigger of mitochondrial Ca2+ mishandling, leading to reduced mitochondrial bioenergetics and cardiac dysfunction.78 Mitochondrial dysfunction in diabetic hearts is further characterised by changes in mitochondrial substrate utilization (i.e. increased reliance on FA-based energy production),79 oxidative stress,80 fragmented mitochondria,81 and impaired mitophagy.82 As opposed to T2DM models,78 hearts of streptozotocin-induced T1DM mice or neonatal murine cardiomyocytes exposed to high glucose levels, display facilitated MAM formation with mitochondrial Ca2+ increase,83 underscoring important myocardial differences in intracellular Ca2+ homeostasis between T1DM and T2DM.

Despite the considerable advances in our mechanistic understanding, a particular concern is that the majority of these results were obtained from rodent cardiomyocytes and there is a clear lack of corresponding observations in human cells.

2.1 Investigating diabetic CM in preclinical models: the importance of clinical confounding factors

The majority of molecular mechanisms in the pathogenesis of diabetic CM has been investigated in rodent models of type 1 or T2DM (Table 1). The models consist of animals with defective insulin actions or signalling, altered cardiac glucose and/or FA utilization, enhanced oxidative stress, and/or cardiac fibrosis.75,84 The most popular animal models include the chemical ablation of the β-cells of the pancreas by streptozotocin, genetic interference with leptin signalling (ob/ob and db/db mice, ZDF rats), the induction of insulin resistance by exposure to high fat diet (HFD) and transgenic animals with a cardiac-specific lipotoxicity.75 However, important confounding factors contributing to cardiac remodelling and dysfunction are rarely considered when dissecting the signalling pathways leading to diabetic CM in rodent models. Experimental approaches that more closely mimic the clinical scenario in T2DM patients are detailed below:

2.1.1 Ageing

Many of the cardiac abnormalities (i.e. increased wall thickness and interstitial myocardial fibrosis, cardiomyocyte hypertrophy) found in diabetic CM are analogous to those induced by ageing.85,86 Recent studies suggest that T2DM accelerates the ageing of the heart and may therefore represent a form of premature senescence leading to premature onset of HF.87 Indeed, T2DM has a dramatic impact on cellular senescence of different types of stem cells, including cardiac stem cells and potentiates the accumulation of senescent cells in the heart.88

Senescent cells display a complex phenotype including DNA damage and genomic instability, ER stress, mitochondria dysfunction, impaired contractile function, hypertrophic growth, and change in gene expression involving a rise of a unique secretory phenotype (senescence-associated secretory phenotype) and induction of senescence-associated beta-galactosidase positivity. Senescent cardiomyocytes secrete growth factors, creating a profibrotic microenvironment and promoting activation of cardiac fibroblasts, which is harmful to the myocardium and triggers processes associated with maladaptive cardiac remodelling.86,89 Furthermore, T2DM induces epigenetic alterations, such as hypermethylation of CpG islands, increased trimethylation of Histone 3 (H3) at lysine (K)4, H3K9, H3K27, and H4K20, as well as a decreased monomethylation and acetylation of H3K9. These epigenetic modifications contribute to senescence through changing the access of transcription factors to promoter/enhancer regions and are complemented by noncoding RNA regulation by microRNA (i.e. miR34a) and long-noncoding RNA.90 For practical and financial reasons, most animal studies make use of animals of young or moderate age. Data is accumulating that aged animals respond differently to perturbations associated with diabetic CM, and we therefore advocate to evaluate the role of aging in relevant models.91

To further underscore the importance of senescence in the pathogenesis of diabetic CM, senolytic drugs have been shown to alleviate myocardial hypertrophy, fibrosis, and diastolic dysfunction in db/db obese mice.92 Such observations have suggested diabetic CM as a model of premature cardiac ageing and that senolytic therapy can prevent this T2DM-related complication.93

2.1.2 Sex

Independent of age, women with T2DM are at higher risk of developing CV diseases (CVD) compared to age-matched men and tend to manifest a more severe cardiac remodelling in diabetic CM.94,95 Interestingly, this sex-dependent aggravation of diabetic CM has been successfully recapitulated in several female rodent models. For instance, ZDF female rats exhibit cardiac hypertrophy with reduced capillary density and increased myocardial structural damage, even though males develop more pronounced fibrosis.96 Increased cardiac hypertrophy and endothelial dysfunction have also been shown in female GK rats compared to males.97 In the db/db mouse model left ventricular pro-hypertrophic and pro-oxidant gene expression were exaggerated in females leading to increased cardiomyocyte size compared to males.98 This difference is probably due to sex hormones and neurohormonal diversity coupled with gender-specific activation of molecular pathways involved in cardiac metabolism/remodelling.99,100 In support of this possibility, animal studies show a sexual dimorphism during the progression of CVD induced by diabetes. Based on the few experimental studies conducted on both sexes of humans and animals, differences in diabetic response seem to be related to relevant sexual dimorphism already present in the non-diabetic state, as demonstrated by differential lipid concentrations and profiles, insulin and glucose control, antioxidant system, nitric oxide (NO) production, energy metabolism, myocardial contractility, and structure.99,101,102 The impact of this sex-dependent effect in diabetes is not fully understood, but differences in metabolic (e.g. glucose, lipid and insulin) control are likely to be pivotal.103 Specifically, in females, the interaction between cardiac insulin and oestrogen signalling, which share common pathways, may modulate many structural and functional features in healthy and diabetic states.103 An illustrative example is the sex-specific dichotomous FA handling pattern: increased accumulation of acylcarnitine (AC) and triglyceride (TG) metabolism with enhanced ROS production in cardiomyocytes has been reported in female GK rats compared to males,104 suggesting a sex-specific FA metabolism and redox biology with potential consequences in diabetic CM. Taken together, considering the equality in prevalence but disparity in clinical presentation, preclinical studies are highly recommended to study both sexes.100,105

2.1.3 Obesity/adiposity

Adipose tissue represents an intersection of pathways involved in longevity, genesis of age-related chronic diseases, metabolic dysfunction, and low-grade inflammation. Obesity and adiposity are causally linked to the development of T2DM and strongly contribute to diabetic CM.106 The cardiac risk of obesity per se, without diabetes and other co-morbid conditions, is underscored by its close association with structural, functional, metabolic, and haemodynamic changes in the heart, leading to a condition clinically termed as obesity CM.107 Obesity CM hearts are characterised by progressive increase in left ventricular (LV) mass, LV remodelling with interstitial fibrosis, and systolic dysfunction that may lead to HF in both patients and rodent models.10,108 Moreover, obesity CM has been recently described as energetic inefficient with reduced ATP delivery in human patients.109 Several wild type high-fat diet (HFD) or genetically modified rodent models have shown to partially recapitulate features of human obesity CM (Table 1). Although conflicting data exists as per the ability of HFD to induce myocardial dysfunction, these are likely explained by differences in mouse strains, the duration and timing of dietary intervention, and composition of diet.109,110 Overall, the mechanisms by which adiposity contributes to cardiac alterations largely overlap with those reported for diabetic CM and include oxidative stress, inflammation, apoptosis, dysregulated autophagy, hypertrophy, interstitial fibrosis, lipotoxicity, and metabolic disturbances. Observations indicate that not only the degree of adiposity counts but the location of body fat accumulation also influences the risk of cardiac dysfunction: ectopic adiposity (visceral, pericardial and epicardial) carries a higher risk than subcutaneous fat,111,112 probably through the release of pro-inflammatory and pro-fibrotic factors.113,114 Interestingly, the effects of HFD on cardiac remodelling seem to be reversible, as a switch from HFD to standard diet for 8 weeks reduced lipid accumulation, myocardial hypertrophy, and fibrosis, and improved myocardial function in 16-week HFD mice.78 These preclinical data are in line with clinical intervention studies, such as gastric bypass, caloric restriction or exercise, intended to reduce myocardial structural and functional consequences of diabetes or obesity.115,116

2.1.4 Sedentary life/exercise

Physical exercise is an important non-pharmacological treatment in T2DM, with high efficacy in delaying or preventing diabetic CM.117 Preclinical studies have identified some mechanisms underlying the exercise-related benefits. Exercise inhibits the pathological processes of myocardial apoptosis, fibrosis, and microvascular alterations through improving myocardial metabolism (improved glucose oxidation and reduced FA oxidation), restoring the physiological regulation of Ca2+ (normalizing depressed expression and function of SERCA2a in HFD + streptozotocin rats) and protecting mitochondrial function.118 Beneficial cardiac effects of exercise are proposed to be mediated by a decrease in adipose tissue senescence with its related pro-fibrotic secretome, independent of improvement in metabolic status in HFD mice.119

2.1.5 Left ventricular pressure overload

Left ventricular pressure overload occurs in a variety of conditions, such as vascular stiffness in advanced age, hypertension, valvular heart disease, often in association with obesity, and diabetes. Its deleterious consequences, i.e. myocardial fibrosis and hypertrophy, are mediated by neurohormonal factors involving the sympathetic nervous system and the renin–angiotensin–aldosterone system (RAAS). The sympathetic nervous system provides the most powerful, but also deleterious, stimulation of cardiac function, via catecholamines and their post-synaptic β-adrenergic receptors (β-AR) including β1-AR, β2-AR, and β3-AR subtypes.120 Interestingly, diabetic CM, similar to other forms of HF, displays alterations of autonomic control with reduction of parasympathetic activity and an increased activity of sympathetic nervous system, which promotes decreased β-AR responsiveness.121–123 The latter increases heart rate, stroke volume, and peripheral vascular resistance and stimulates the RAAS, exacerbating left ventricular dysfunction. At the molecular level, elevated sympathetic drive enhances β1-AR signalling, which promotes hypertrophy, interstitial fibrosis, cardiomyocyte apoptosis and impairs energy metabolism and myocardial function.120 Additional mechanistic studies suggested that a canonical downstream effector of β-AR, the cyclic AMP-dependent protein kinase A (PKA) may be involved in the deficient ventricular performance and metabolism in the mouse diabetic heart,124 potentially giving way to other cAMP effectors, such as the Epac proteins.125 Interestingly, the relationship between insulin resistance and β-AR signalling is emerging as an important focal node in the pathogenesis of diabetic CM since hyperinsulinemia may play a role in desensitization of β-AR signalling in T2DM. This is well illustrated in a study showing that in a diabetic CM murine model induced by HFD, myocardial injury and dysfunction could be reversed by pharmacologically inhibition of β2-AR or G protein-coupled receptor kinase 2 activity.126 In contrast to cardiac β1- and β2-AR, the role of the β3-AR in the diabetic heart has been hardly investigated. It was reported that a β3-adrenoceptor-mediated negative inotropic effect contributes to the altered positive inotropic response induced by β-adrenoceptor activation in diabetic rat heart.127

2.1.6 Chronic intermittent hypoxia

Prevalence and severity of OSA is higher among diabetic individuals compared to non-diabetic subjects.128 OSA is associated with metabolic and CV co-morbidities including hypertension, arrhythmia, stroke, coronary heart disease, which supports OSA as a major health burden. Mechanistic studies in rodents subjected to chronic intermittent hypoxia (the pathophysiologic basis of OSA) found that OSA-induced CV dysfunction (vascular remodelling, endothelial dysfunction, early atherosclerosis and increased arterial blood pressure) depends on oxidative stress- and HIF1α-driven sympathetic overactivity. Specifically, increased levels of ROS and HIFα activate chemoreflex and suppress baroreflex, thereby stimulating the sympathetic nervous system, increasing LV afterload, and contributing to insulin resistance and T2DM.129

2.2 Crosstalk between metabolic organs and the heart beyond cardiac glucose toxicity

To date, there is a need to consider each major organ, i.e. heart, liver, adipose tissue, skeletal muscle, lung, kidney, and brain operating as an integrated network within the human body in response to dysregulated metabolism. In particular, T2DM is associated with progressive microvascular disorders and systemic inflammatory processes, inducing fibrosis in several organs, including the CV system, liver, adipose tissue, kidney, and skeletal muscle. During the progression of these fibro-inflammatory processes, there are significant haemodynamic and metabolic interactions between these organs, which need to be assessed to predict health trajectories in T2DM patients,130 and more specifically the progression towards diabetic CM and HF with preserved ejection fraction (HFpEF).131,132 Indeed, dysregulation of both the immune system and microcirculation through endothelial cell dysfunction and procoagulant changes contributes to diabetic CM beyond hyperglycaemia, insulin resistance, and metabolic derangements.74,130 Importantly, the microcirculation impacts on insulin sensitivity by affecting the delivery of insulin and glucose to skeletal muscle. Thus, endothelial dysfunction and extracellular matrix remodelling promote the progression from prediabetes to diabetes and the development of diabetic CM and other T2DM complications, including HFpEF and chronic kidney disease (CKD).133,134 Thus, whilst HFpEF was initially considered as a disorder characterized by hypertension, cardiac hypertrophy, and diastolic dysfunction, the pandemics of obesity and T2DM have modified the HFpEF syndrome. As a result, HFpEF is now recognized as a multisystem disorder involving the heart, lungs, kidneys, skeletal muscle, adipose tissue, vascular system, and immune and inflammatory signalling.131,135 We acknowledge that in clinical practice, HFpEF and diabetic CM are often difficult to distinguish. HFpEF can be the result of a large number of triggers, including diabetes, whereas diabetic CM refers to myocardial structural abnormalities that are predominantly caused by diabetes (Table 2).

Table 2

Comparison between diabetic cardiomyopathy and heart failure with preserved ejection fraction

Clinical presentation or factorDCMHFpEF
DiabetesMandatoryVery common (>50%)
HFpEFCommon, but also be HF(m)rEFMandatory
AgeElderly(>60 years)Very elderly (>75 years)
Sex distribution50:50%Female dominance(∼70%)
HypertensionVery commonVery common
Coronary artery disease (CAD)No obstructive CADObstructive CAD common (30%)
ObesityVery common (>80%)Common (>50%)
Diastolic LV dysfunctionBy defaultCommon
Myocardial metabolismSignificantly altered
Favouring FA over glucose
Lipotoxity
Ketone utilization
Usually altered Switch from FA to glucose Lipotoxicity
Mitochrondial dysfunction and lower biogenesis+++++
Autonomic neuropathy++
Fibrosis+++
Clinical presentation or factorDCMHFpEF
DiabetesMandatoryVery common (>50%)
HFpEFCommon, but also be HF(m)rEFMandatory
AgeElderly(>60 years)Very elderly (>75 years)
Sex distribution50:50%Female dominance(∼70%)
HypertensionVery commonVery common
Coronary artery disease (CAD)No obstructive CADObstructive CAD common (30%)
ObesityVery common (>80%)Common (>50%)
Diastolic LV dysfunctionBy defaultCommon
Myocardial metabolismSignificantly altered
Favouring FA over glucose
Lipotoxity
Ketone utilization
Usually altered Switch from FA to glucose Lipotoxicity
Mitochrondial dysfunction and lower biogenesis+++++
Autonomic neuropathy++
Fibrosis+++

DCM, diabetic cardiomyopathy; HFpEF, heart failure with preserved ejection fraction; LV, left ventricular.

Table 2

Comparison between diabetic cardiomyopathy and heart failure with preserved ejection fraction

Clinical presentation or factorDCMHFpEF
DiabetesMandatoryVery common (>50%)
HFpEFCommon, but also be HF(m)rEFMandatory
AgeElderly(>60 years)Very elderly (>75 years)
Sex distribution50:50%Female dominance(∼70%)
HypertensionVery commonVery common
Coronary artery disease (CAD)No obstructive CADObstructive CAD common (30%)
ObesityVery common (>80%)Common (>50%)
Diastolic LV dysfunctionBy defaultCommon
Myocardial metabolismSignificantly altered
Favouring FA over glucose
Lipotoxity
Ketone utilization
Usually altered Switch from FA to glucose Lipotoxicity
Mitochrondial dysfunction and lower biogenesis+++++
Autonomic neuropathy++
Fibrosis+++
Clinical presentation or factorDCMHFpEF
DiabetesMandatoryVery common (>50%)
HFpEFCommon, but also be HF(m)rEFMandatory
AgeElderly(>60 years)Very elderly (>75 years)
Sex distribution50:50%Female dominance(∼70%)
HypertensionVery commonVery common
Coronary artery disease (CAD)No obstructive CADObstructive CAD common (30%)
ObesityVery common (>80%)Common (>50%)
Diastolic LV dysfunctionBy defaultCommon
Myocardial metabolismSignificantly altered
Favouring FA over glucose
Lipotoxity
Ketone utilization
Usually altered Switch from FA to glucose Lipotoxicity
Mitochrondial dysfunction and lower biogenesis+++++
Autonomic neuropathy++
Fibrosis+++

DCM, diabetic cardiomyopathy; HFpEF, heart failure with preserved ejection fraction; LV, left ventricular.

Table 3

Subcellular hallmarks in major experimental rodent models of diabetic cardiomyopathy

MICERATS
HFDTransgenicT1DMT2DMT1DMT2DM
C57BL6ob/obMHC-PPARαSTZdb/dbSTZZDFGK
Fatty acid oxidation
Glucose oxidation
Lipotoxicity
Oxidative stress
InflammationN/A
ApoptosisN/A
Calcium handling↑/=
Mitochondrial function↓/=
Molecular changes (expression/activity)↑CD36Leptin mutation↑Cardiac PPARα ↑CD36↑NADPH oxidase (subunit 47) ↑PPARα ↑creatinine kinase ↓miR-133 ↑miR-195Leptin receptor mutation↑NLRP3↑SERCA
References10,83,84, 220–22784,220,221,228–23222,84,23322,84,220,234–24384,220,232,244–24884,222,249–25484,220,221,255–26184,237,262–269
MICERATS
HFDTransgenicT1DMT2DMT1DMT2DM
C57BL6ob/obMHC-PPARαSTZdb/dbSTZZDFGK
Fatty acid oxidation
Glucose oxidation
Lipotoxicity
Oxidative stress
InflammationN/A
ApoptosisN/A
Calcium handling↑/=
Mitochondrial function↓/=
Molecular changes (expression/activity)↑CD36Leptin mutation↑Cardiac PPARα ↑CD36↑NADPH oxidase (subunit 47) ↑PPARα ↑creatinine kinase ↓miR-133 ↑miR-195Leptin receptor mutation↑NLRP3↑SERCA
References10,83,84, 220–22784,220,221,228–23222,84,23322,84,220,234–24384,220,232,244–24884,222,249–25484,220,221,255–26184,237,262–269

T1DM, type 1 diabetes mellitus; T2DM, type 2 diabetes mellitus; HFD, high-fat diet; GK, Goto-Kakizaki rats; MHC-PPARα, mice with cardiomyocyte-specific overexpression of peroxisome proliferator activated receptor α (PPARα), STZ: streptozotocin; ZDF, Zucker diabetic fatty rats; SD, Sprague-Dawley; miR, microRNA; N/A, data not available.

Table 3

Subcellular hallmarks in major experimental rodent models of diabetic cardiomyopathy

MICERATS
HFDTransgenicT1DMT2DMT1DMT2DM
C57BL6ob/obMHC-PPARαSTZdb/dbSTZZDFGK
Fatty acid oxidation
Glucose oxidation
Lipotoxicity
Oxidative stress
InflammationN/A
ApoptosisN/A
Calcium handling↑/=
Mitochondrial function↓/=
Molecular changes (expression/activity)↑CD36Leptin mutation↑Cardiac PPARα ↑CD36↑NADPH oxidase (subunit 47) ↑PPARα ↑creatinine kinase ↓miR-133 ↑miR-195Leptin receptor mutation↑NLRP3↑SERCA
References10,83,84, 220–22784,220,221,228–23222,84,23322,84,220,234–24384,220,232,244–24884,222,249–25484,220,221,255–26184,237,262–269
MICERATS
HFDTransgenicT1DMT2DMT1DMT2DM
C57BL6ob/obMHC-PPARαSTZdb/dbSTZZDFGK
Fatty acid oxidation
Glucose oxidation
Lipotoxicity
Oxidative stress
InflammationN/A
ApoptosisN/A
Calcium handling↑/=
Mitochondrial function↓/=
Molecular changes (expression/activity)↑CD36Leptin mutation↑Cardiac PPARα ↑CD36↑NADPH oxidase (subunit 47) ↑PPARα ↑creatinine kinase ↓miR-133 ↑miR-195Leptin receptor mutation↑NLRP3↑SERCA
References10,83,84, 220–22784,220,221,228–23222,84,23322,84,220,234–24384,220,232,244–24884,222,249–25484,220,221,255–26184,237,262–269

T1DM, type 1 diabetes mellitus; T2DM, type 2 diabetes mellitus; HFD, high-fat diet; GK, Goto-Kakizaki rats; MHC-PPARα, mice with cardiomyocyte-specific overexpression of peroxisome proliferator activated receptor α (PPARα), STZ: streptozotocin; ZDF, Zucker diabetic fatty rats; SD, Sprague-Dawley; miR, microRNA; N/A, data not available.

How interorgan crosstalk during T2DM specifically contributes to diabetic CM requires further exploration. Animal models of T2DM represent a unique approach to test the mechanisms of such organ interactions and to assess how the pathological state developing in one organ, can lead to deleterious functional and structural consequences in the heart (Table 3).

2.2.1 Kidney

T2DM is directly related to both CKD and CVD. Patients with diabetes and CVD are twice as likely to develop CKD than those without CVD.133–136 Furthermore, the co-incidence of HFpEF and CKD is very strong since approximately 50% of the patients with HFpEF also suffer from CKD.132,137 To further underscore a pathophysiological crosstalk, activated inflammatory cascades and endothelial dysfunction in renal injury promote features of HFpEF, such as cardiomyocyte stiffening and myocardial fibrosis.132 Beside clinical evidence, causal relationship between CKD and HFpEF came from an experimental rat model of CKD induced by nephrectomy, which resulted in a cardiac HFpEF-like phenotype, with left ventricular hypertrophy and diastolic dysfunction.138 The kidney-heart relationship is also achieved by complex interactions involving neurohormonal pathways.139 This is well illustrated with the RAAS system, which is overactivated during CKD and causes a cascade of events leading to vasoconstriction, increased sodium retention, and reduced water excretion. All of which increase blood volume expansion and restore perfusion pressure and therefore may contribute to the development of HFpEF.139 Beside its renal effects, aldosterone directly promotes cardiac fibrosis, left ventricular hypertrophy, and coronary microvascular dysfunction.135 Additional renal factors such as uremic toxins and galectin 3 may also have a direct impact on the heart and/or coronary microvasculature and therefore may play a role in the pathogenesis of HFpEF.140,141 Finally, in indirect support of a kidney involvement, large clinical studies established that whereas tight glycaemic control alone does not,65–69 but GLP-1 receptor agonists142 and SGLT-2 inhibitors,143,144 which also improved kidney disease,142,144,145 lowered the risk for HF in diabetic patients. Thus, these important interactions between T2DM, renal dysfunction and diabetic CM seem to induce a downward spiral of deleterious events, whose interruption represents a novel therapeutic opportunity.146

2.2.2 Adipose tissue

A growing body of evidence supports the existence of a two-way adipose-myocardial axis in which products released from fat affect myocardial metabolism and function, whilst peptides secreted from the heart affect FA disposal. Accumulation of ectopic fat in various organs, e.g. in heart, liver, pancreas and kidney has been identified as an important marker in the pathogenesis of T2DM in both human and animal studies.147–149 Although the causal relationship between the pathophysiological status of white adipose tissue and cardiac lipotoxicity remains elusive, elevated lipolytic rate in adipose tissue has been demonstrated to contribute to the overall augmentation of plasma lipid levels, as observed in the majority of patients suffering from HF. Excessive release of FA from adipose tissue contributes to myocardial insulin resistance with subsequent metabolic inflexibility characterised by a shift in cardiac energy expenditure towards a near-exclusive and less oxygen-efficient FA oxidation. The perpetuation of this metabolic deregulation leads to the development of cardiac lipotoxicity.150,151 Cardiac lipid overload promotes the formation of cytotoxic intermediates (diacyl-glycerols and ceramides) and enhances ROS generation through exacerbated peroxisomal and mitochondria FA oxidation. Both intermediate lipotoxic species and ROS affect mitochondrial function and Ca2+-handling proteins promoting cardiac dysfunction.152,153

Adipose tissue is an important source of inflammatory mediators (Tumor Necrosis Factor: TNF-α, Interleukin 6: IL6, Interleukin 8: IL-8, Monocyte chemoattractant protein-1: MCP-1) and adipokines (leptin, resistin, and omentin), which may act in an autocrine, paracrine, and endocrine manner, ultimately furthering cardiac injury.114,154 In contrast, the anti-inflammatory adipokine, adiponectin is inversely correlated with myocardial adiposity. Whilst visceral adipose tissue contributes to a low-level and sustained systemic inflammation, pericardial and epicardial fat can directly affect the underlying myocardium by local diffusion of secreted inflammatory mediators.155–157 Another ectopic fat source known to influence the heart is the perivascular adipose tissue (PVAT), whose volume increases proportionally to visceral adipose tissue.158 In obesity, PVAT has been shown to shift from an anti-inflammatory and vasodilatory profile towards a proinflammatory status with impaired vasodilation favouring the progression of vascular disease.159,160

Finally, a prominent role for atrial and B-type natriuretic peptides (ANP and BNP, respectively) has been proposed in the crosstalk between the heart and the adipose tissue.161 As such, the induction of lipolysis by natriuretic peptides secreted by the damaged heart has been suggested to counteract obesity, with a disproportionately greater effect in reducing visceral adipose tissue than subcutaneous adipose tissue.162 On the other hand, increased release of adipocyte FA may contribute to cardiac steatosis and cardiac cachexia.163

Further analysis of the crosstalk between adipose tissue and the heart may identify new treatment options, such as targeting lipolysis and cardiac lipid metabolism in diabetic CM to avoid its progression towards HFpEF.

2.2.3 Liver

Several studies support the bidirectional crosstalk between the heart and liver and the consequences of simultaneous development of hepatic metabolic diseases, diabetic CM, and HF. A better understanding of this hepato-cardiac axis is required to ensure an effective management of T2DM patients with heart or liver diseases in order to improve overall prognosis.

Whilst T2DM and metabolic diseases (obesity and non-alcoholic fatty liver disease) are important risk factors to induce cardiac dysfunction,108,164 a growing body of evidence suggests that the dysfunctional heart per se could affect both systemic metabolism and liver function, and thus, create a vicious injurious cycle between heart and liver. The close association among cardiac and metabolic diseases suggests a common pathophysiological basis. Notably, in metabolic diseases, the heart and liver share similar intracellular defects such as mitochondrial dysfunction, ER stress, lipotoxicity, and disrupted Ca2+ homeostasis. Interestingly, MAM exchange phospholipids and Ca2+ as well as regulate metabolic homeostasis and signalling.165 Of note, reduction of ER-mitochondria communication was observed in both heart78 and liver166 of HFD mice. In the heart, decreased ER-mitochondria communication caused mitochondrial dysfunction leading to diabetic CM, whereas in the liver, disrupted ER-mitochondria interactions undermine hepatic metabolic flexibility and insulin sensitivity. Therefore, targeting MAM could be a new strategy to concomitantly improve both heart and liver function in T2DM.

In addition, the heart secretes proteins referred to as cardiokines, which go beyond local cardiac effects, and mediate changes in extracardiac tissues, including liver function. For example, the cardiac ANP attenuates glycolysis and increases gluconeogenesis in rat liver.167 Other studies showed that the heart controls systemic metabolism via the cardiac-specific microRNA-208a and the mediator complex subunit 13 (MED13) signalling in rodent cardiomyocytes.168 Overexpression of MED13 or inhibition of miR-208a in cardiac tissue of transgenic mice enhanced lipid uptake, β-oxidation, mitochondrial content, and other genes involved in FA utilization in adipose tissue and liver,169 supporting the existence of a functionally relevant, metabolic crosstalk between the heart and liver.

2.2.4 Skeletal muscle

Sarcopenia is characterized by a loss of skeletal muscle strength due to reduction in the quality and quantity of muscle mass, replacement of myofibers with fat, changes in muscle metabolism, oxidative stress, degeneration of neuromuscular junctions, and increased fibrosis. Whilst sarcopenia has been described in elderly individuals, mounting evidence suggests a higher prevalence in T2DM patients. Sarcopenia in T2DM patients may be caused by different mechanisms, such as impaired insulin sensitivity, chronic hyperglycaemia, advanced glycosylation end products, subclinical inflammation, microvascular, and macrovascular complications.170 It seems that the opposite also applies; patients with sarcopenia are at increased risk to develop T2DM due to reduced organismal capacity to catabolise high-energy nutrients.170,171 In addition, sarcopenia is associated with CVD172,173 and both share common risk factors, such as altered glucose metabolism, insulin resistance, inflammation, and metabolic syndrome.174 For instance, T2DM patients with chronic HF exhibit severe skeletal muscle fibre atrophy, capillary remodelling and impaired mitochondrial function, characterized by mitochondrial complex I dysfunction with ROS overproduction.175

2.2.5 Brain

Recent studies shed light on the relationship between the brain and CV system, and how the brain-heart axis regulates T2DM.176 Cohort studies highlight the link between Alzheimer’s disease and T2DM,177 whilst drugs that are currently approved for the treatment of T2DM, such as metformin, have shown promising results in improving cognitive function, and even preventing the development of Alzheimer’s disease in diabetic patients.178

2.3 Investigating diabetic CM in preclinical models: the role of cellular crosstalk within the heart

In analogy to interorgan signalling, adjacent cells also communicate in a paracrine and autocrine fashion, where a given cell can detrimentally affect neighbouring cells, leading to a vicious cycle and subsequent cardiac dysfunction. In addition to its signature parenchymal cells, the contracting cardiomyocytes, the heart contains many other cells, including fibroblasts, smooth muscle cells, endothelial cells, and resident macrophages. Healthy crosstalk between these different cells ensures myocardial homeostasis, but a pathologically altered cell-cell communication may initiate and propagate adverse cardiac remodelling leading to the development of diabetic CM.135,179

2.3.1 Fibroblasts/cardiomyocytes

Cardiac fibroblasts play a crucial role in extracellular matrix (ECM) turnover, as they are involved in both synthesis and degradation of ECM components through matrix metalloproteinasesand tissue inhibitors of metalloproteinases. Fibroblasts adhere to ECM proteins through integrins that are critical mediators of cell attachment, adhesive signalling, and remodelling of collagen fibrils. Excessive cardiac ECM deposition is a key feature of the remodelling response in diabetic CM and promotes myocardial stiffness and cardiac dysfunction in rodent models of diabetes.180 Experimentally, high glucose levels induce cardiac fibroblasts into a state of increased proliferation,181 with increased DNA and collagen synthesis as well as fibronectin and TGF-beta-1 gene expression.182,183 Genetic inhibition of α11β1 integrin in STZ diabetic mice prevents the progression of fibrosis and abnormal cardiomyocyte growth, indicating that this specific integrin plays a critical role in modifying fibroblast-cardiomyocyte-ECM interactions.184 Crosstalk between cardiomyocytes and fibroblasts is also associated with a cardiomyocyte switch to a fibrogenic phenotype, characterized by increased synthesis and release of cytokines that induce fibroblast proliferation and activation, as well as proinflammatory molecules that trigger fibrosis through activation of immune cells.180

2.3.2 Endothelial cells

Diabetic CM is associated with coronary microvascular dysfunction, which impairs coronary blood flow and myocardial perfusion.185 Abnormalities in the coronary microcirculation result from endothelial cell dysfunction, which is considered a central mechanism in HFpEF pathophysiology.8,160 Indeed, endothelial cells have altered paracrine signalling to cardiomyocytes by reducing the bioavailability of vasodilator molecules, NO and endothelium-derived hyperpolarizing factors (EDHFs), thereby limiting blood flow and promoting leukocyte infiltration in the myocardium.186 The latter leads to activation of myofibroblasts and interstitial collagen deposition. As part of T2DM-associated glucotoxicity and lipotoxicity, endothelial cells generate ROS and reactive nitrogen species (RNS) that uncouple endothelial NO synthase (eNOS) activity (by oxidizing eNOS cofactor tetrahydrobiopterin) leading to decreased NO bioavailability.187 This effect together with insulin resistance converges on and minimizes the activity of guanylate cyclase and cyclic guanosine monophosphate- protein kinase G signalling that results in deranged titin phosphorylation and increased cardiomyocyte hypertrophy, exacerbating wall stiffness in diabetic hearts.188 Endothelial cells can also contribute to the development of cardiac fibrosis through endothelial-to-mesenchymal transition to myofibroblasts.189

2.3.3 Immune cells

Numerous experimental and clinical studies have reported a role of adaptive immunity in diabetic CM pathogenesis.190–192 T2DM is associated with chronic systemic inflammation, which leads to leukocyte activation and recruitment to various organs, further aggravating inflammatory cardiac tissue remodelling over time.191 This chain of events results in cardiac fibrosis as resident fibroblasts become activated in response to pathophysiologic conditions, which for the heart, leads to wall stiffening and decreased contractility.180 Although the role of B cells is still unclear, T cell-derived immune response has been shown to contribute to the progression of diabetic CM.192 In particular, in STZ-induced rodent models of diabetic CM, increased infiltration of T lymphocytes into the myocardium is positively correlated with increased collagen deposition and wall stiffness,191 whilst genetic depletion of CD4+ T cells protects against cardiac fibrosis and impairment in LV function.192,193 Yet, recent studies have further delineated the contribution of each T-lymphocyte subset in diabetic CM. Proinflammatory T helper cells Th1, Th17, and Th22 subtypes are increased in diabetic CM,194 whereas the activation of anti-inflammatory Th2 and Foxp3+ Treg subtypes is delayed or impaired,195 overall promoting chronic inflammatory tissue damage. Increased neutrophil/lymphocyte ratio (an indicator of systemic inflammation) is associated with the occurrence of subclinical diabetic CM.196 As per potential mechanisms involved, the sphingosine-1-phosphate (S1P)/S1P-receptor signalling axis regulating T cell trafficking, activation, and polarization may be of importance.197 Indeed, targeted deletion of T-cell S1P-R or administration of fingolimod (an S1P-receptor modulator) both reduce myocardial fibrosis and improve cardiac function in STZ-induced diabetic CM mice.192,198

Macrophages also play a key role in regulating inflammatory responses and homeostatic maintenance of the myocardium. Normally in injured tissue, efferocytosis allows macrophages to engulf apoptotic cells and cellular debris to reduce inflammation.199 Efferocytosis is regulated by many processes in high-glucose milieu. In particular, the metalloproteinase disintegrin and metalloproteinase domain-containing protein 9 (ADAM-9) were shown to be upregulated in macrophages, secondary to a downregulation of miR-126, which increased MER proto-oncogene, tyrosine kinase (MerTK) cleavage with a net effect of reduced efferocytosis.200 Interestingly, human diabetic hearts display the same molecular signatures in terms of miR-126, ADAM9, and cleaved MerTK expression, suggesting that this pathway may be involved in regulating human diabetic CM progression. Recently, cardiac-resident MHCIIhigh macrophages showed a pathogenic role in cardiac remodelling through production of IL-10. The profibrotic effect of IL-10 autocrine loop promotes macrophages to secrete osteopontin and TGFβ, which induce cardiac fibroblasts into producing collagen that results in cardiac fibrosis with increased cardiac stiffness.201 Therefore, a new understanding of communication between cardiac macrophages and fibroblasts could lead to novel therapeutic strategies for diabetic CM and its progression towards HF.

2.4 Investigating diabetic CM in preclinical models: identifying new biomarkers and therapeutic targets

2.4.1 Biomarkers

Since T2DM patients at high risk of developing HF display altered metabolism in cardiomyocytes, with underlying changes in protein and metabolite profiles related hyperglycaemia, lipotoxicity and oxidative stress, a systems biology approach may identify a specific signature of diabetic CM. To reduce disease burden, it is imperative to develop non-invasive biomarkers to detect and characterize diabetic CM processes at their early and possibly reversible stages in order to reveal new therapeutic targets and to follow disease progress. These last years, new methods have emerged, which offer a great potential to identify such biomarkers. Big datasets derived from in silico predictive models, imaging, and OMICS technologies (metabolomics, lipidomics, transcriptomics, proteomics) may be used for developing multiparametric datasets to assist improved diagnostic and therapeutic decisions.

Metabolic alterations and insulin resistance are early signs of future cardiac dysfunction and have a causative role in the development of diabetic CM.202 Metabolomics using different analytical techniques such as magnetic resonance spectroscopy, mass spectrometry and chromatography203 are powerful approaches to follow simultaneous changes in multiple metabolite levels occurring in the diabetic heart. Indeed, cardiac energetic metabolism assessed by the PCr/ATP ratio, is reduced in some studies,204 although some discrepancies exist depending on the models.205,206 In parallel, lipid metabolism is altered with increased FA oxidation and lipid accumulation.147

In silico predictive methods have the potential to reveal or to confirm effective biomarkers. Using this approach and exploiting meta-analysis of transcriptomic datasets, differential expression levels of lysyl oxidase like 2 (LOXL2) and electron transfer flavoprotein beta subunit (ETFβ) in serum and heart tissue of 6–16-week-old db/db mice correlated closely with a reduced LV diastolic dysfunction, supporting the use of LOXL2 and ETFβ as early predictive biomarkers for diabetic CM.207

Moreover, systematic multiorgan biobanking of porcine models of diabetes and obesity subjected to molecular profiling by transcriptomics, proteomics and metabolomics has been proposed to better understand tissue-specific pathogenic mechanisms and organ crosstalk with the prospect of revealing novel molecular targets.208

In the field of imaging technologies, the development of machine learning algorithms aims to provide more accurate biomarkers.209 Thus, combining imaging, radiomics and multi-OMICS data with machine learning will provide large datasets of parameters allowing to find biomarkers for early diagnosis and monitoring progress of diabetic CM.

2.4.2 New therapeutic targets

Drug development is time-consuming and costly, urging the use of precision medicine to replace the ‘one size fits all’ paradigm with more patient tailoring approaches. Understanding T2DM-specific mechanisms shall lead to opportunities of developing better therapies. Mechanistic studies have demonstrated dramatic glucotoxicity in the heart, and linked it to accelerated sugar-related protein modifications, such as O-GlcNAcylation210 and AGE formation,211 as well as increased ROS formation.212 Yet, most interventional studies focusing on the reduction of plasma glucose in T2DM patients found at most modest improvement213 and even deleterious effects in HF outcomes.66–69 To underscore the importance of alternative mechanisms, recent benefits obtained with SGLT2 inhibitor (SGLT2i) treatment for HF and CKD were partly independent of their hypoglycaemic effects.143,144 Many potential mechanisms have been proposed for SGLT2i.214–216 For example, it has been suggested that a nephroprotective effect with natriuresis, diuresis and decreased blood volume that reduced preload and afterload are possible mechanisms.143,144 Reduced cardiac oxidative stress and fibrosis have also been observed with SGLT2i treatment.217 Occurrence of ketoacidosis prompted the idea that plasma ketone bodies may serve as an alternative and efficient source of cardiac fuel.218 It has also been proposed that SGLT2 inhibitors have off-target pharmacology by directly inhibiting cardiac NHE1 activity and protect the myocardium under ischaemic conditions.219 Nevertheless, confirmation of SGLT2i’s specific cardioprotective mechanisms remains elusive to date.

Research efforts need therefore to focus on finding therapeutic strategies to inhibit pathophysiological pathways and reduce the risk of diabetic CM. In addition, the understanding of diabetic CM pathophysiology should generate awareness regarding its multiorgan nature. Thus, holistic approaches considering the complexity of myocardial damage induced by T2DM along with the functional interplay between different key organs will advance our knowledge of diabetic CM. This type of multidimensional approach will increase the likelihood of early diagnosis and the translational success of new drugs in development. Currently, there are no specific therapies for diabetic CM. Further refinement of diabetic CM molecular signatures derived from improved preclinical models should provide new mechanistic insights leading to specific targets, drugs, biomarkers, and effective patient management in the future.

3. Future perspectives

Animal models have provided valuable insight into the initiation and progression of diabetic CM, including the revelation of some underlying molecular mechanisms. In addition, they are irreplaceable for testing new treatments and identifying possible side-effects. Despite these undeniable virtues, experimental models have failed to reproduce all structural, functional, and molecular alterations of human diabetic CM, posing as one of the obstacles to advance patient care. An additional intriguing issue with pathophysiological relevance is the failure of antidiabetic drugs to combat diabetic CM. Given the myriads of confounding factors in clinical reality, it is probably impossible to propose a single best model of rodent diabetic CM recapitulating human diabetic CM in its entirety. Indeed, diabetic patients experiencing various additional ‘stresses’ (advanced liver or kidney disease, sarcopenia, OSA, Alzheimer’s disease, etc.) may therefore develop different versions of diabetic CM, hence would mostly benefit from tailored therapeutic interventions. Thus, the design of rodent models for studies on diabetic CM is complex and should involve commonly coexisting comorbidities in humans to reflect specific endotypes. Such new models may include but do not limit to middle-aged or old animals (10–12 months or and ∼ 2 years of age) fed with HFD and undergoing intermittent hypoxemia, or HFD model with experimental renal impairment (i.e. 1K1C, 2K1C), or HFD + carbon tetrachloride to induce additional liver injury to cover the full spectrum of NAFLD. Models also need optimizing in terms of duration and composition (e.g. Omega 3/Omega 6 ratio) of HFD, use of rodents with different ages, both genders, etc. We believe that the development of endotype-specific models will be the preclinical response to personalised medicine, facilitating the discovery of new targets and translation to bedside.’

Acknowledgements

All authors contributed substantially to drafting and revising the manuscript. F.L. and L.B. equally contributed to the work as co-first authors.

This manuscript is submitted on behalf of CARDIATEAM consortium which has approved the submission of this article: ALASSAD Lara, ASSELBERGS Folkert, AUDUREAU Etienne, BERGEROT Cyrille, BERMEJO Javier, BEULENS Joline W.J., BOITARD Christian, DEUX Jean-François, DEVAUX Yvan, D'HOOGE Jan, DUTOUR Anne, FREITAG Daniel, FRESE Karen, GABORIT Bénédicte, GAUTIER Jean François, HANDOKO M Louis, HEYMANS Stephane, IBBERSON Mark, JACOBS Bart, JULLA Jean-Baptiste, KORNERUP Kristin, LANG Chim C, LARGER Etienne, LIECHTI Robin, LUCIANI Alain, MARX Nikolaus, MATULLO Giuseppe, MEDER Benjamin, MEYER Jutta, MIREA Oana, MULLER-WIELAND Dirk, OERLEMANS Marish, PIZARD Anne, PREVOST Sonia, REICH Christoph, SAM Flora, TACHER Vania, THIBAULT Helene, TRUCCO Emanuele, VAN EMPEL Vanessa, WANG-SATTLER Rui.

Funding

This work was supported by the Innovative Medicines Initiative (H2020, IMI), (Grant number: 821508 to G.A.D.).

References

1

Cosentino
F
,
Grant
PJ
,
Aboyans
V
,
Bailey
CJ
,
Ceriello
A
,
Delgado
V
,
Federici
M
,
Filippatos
G
,
Grobbee
DE
,
Hansen
TB
,
Huikuri
HV
,
Johansson
I
,
Juni
P
,
Lettino
M
,
Marx
N
,
Mellbin
LG
,
Ostgren
CJ
,
Rocca
B
,
Roffi
M
,
Sattar
N
,
Seferovic
PM
,
Sousa-Uva
M
,
Valensi
P
,
Wheeler
DC
,
ESC Scientific Document Group
.
2020 ESC guidelines on diabetes, pre-diabetes, and cardiovascular diseases developed in collaboration with the EASD
.
Eur Heart J
2019
;
41
:
255
323
.

2

Kannel
WB
,
Hjortland
M
,
Castelli
WP
.
Role of diabetes in congestive heart failure: the Framingham study
.
Am J Cardiol
1974
;
34
:
29
34
.

3

Rubler
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
.

4

Seferovic
PM
,
Petrie
MC
,
Filippatos
GS
,
Anker
SD
,
Rosano
G
,
Bauersachs
J
,
Paulus
WJ
,
Komajda
M
,
Cosentino
F
,
de Boer
RA
,
Farmakis
D
,
Doehner
W
,
Lambrinou
E
,
Lopatin
Y
,
Piepoli
MF
,
Theodorakis
MJ
,
Wiggers
H
,
Lekakis
J
,
Mebazaa
A
,
Mamas
MA
,
Tschope
C
,
Hoes
AW
,
Seferovic
JP
,
Logue
J
,
McDonagh
T
,
Riley
JP
,
Milinkovic
I
,
Polovina
M
,
van Veldhuisen
DJ
,
Lainscak
M
,
Maggioni
AP
,
Ruschitzka
F
,
McMurray
JJV
.
Type 2 diabetes mellitus and heart failure: a position statement from the heart failure association of the European Society of Cardiology
.
Eur J Heart Fail
2018
;
20
:
853
872
.

5

Kishi
S
,
Gidding
SS
,
Reis
JP
,
Colangelo
LA
,
Venkatesh
BA
,
Armstrong
AC
,
Isogawa
A
,
Lewis
CE
,
Wu
C
,
Jacobs
DR
Jr
,
Liu
K
,
Lima
JA
.
Association of insulin resistance and glycemic metabolic abnormalities with lv structure and function in middle age: the CARDIA study
.
JACC Cardiovasc Imaging
.
2017
;
10
:
105
114
.

6

Ernande
L
,
Thibault
H
,
Bergerot
C
,
Moulin
P
,
Wen
H
,
Derumeaux
G
,
Croisille
P
.
Systolic myocardial dysfunction in patients with type 2 diabetes mellitus: identification at MR imaging with cine displacement encoding with stimulated echoes
.
Radiology
2012
;
265
:
402
409
.

7

Ernande
L
,
Bergerot
C
,
Girerd
N
,
Thibault
H
,
Davidsen
ES
,
Gautier Pignon-Blanc
P
,
Amaz
C
,
Croisille
P
,
De Buyzere
ML
,
Rietzschel
ER
,
Gillebert
TC
,
Moulin
P
,
Altman
M
,
Derumeaux
G
.
Longitudinal myocardial strain alteration is associated with left ventricular remodeling in asymptomatic patients with type 2 diabetes mellitus
.
J Am Soc Echocardiogr
2014
;
27
:
479
488
.

8

Seferovic
PM
,
Paulus
WJ
.
Clinical diabetic cardiomyopathy: a two-faced disease with restrictive and dilated phenotypes
.
Eur Heart J
2015
;
36
:
1718
1727
.
1727a-1727c
.

9

Marwick
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
.

10

Ternacle
J
,
Wan
F
,
Sawaki
D
,
Surenaud
M
,
Pini
M
,
Mercedes
R
,
Ernande
L
,
Audureau
E
,
Dubois-Rande
JL
,
Adnot
S
,
Hue
S
,
Czibik
G
,
Derumeaux
G
.
Short-term high-fat diet compromises myocardial function: a radial strain rate imaging study
.
Eur Heart J Cardiovasc Imaging
2017
;
18
:
1283
1291
.

11

Raher
MJ
,
Thibault
HB
,
Buys
ES
,
Kuruppu
D
,
Shimizu
N
,
Brownell
AL
,
Blake
SL
,
Rieusset
J
,
Kaneki
M
,
Derumeaux
G
,
Picard
MH
,
Bloch
KD
,
Scherrer-Crosbie
M
.
A short duration of high-fat diet induces insulin resistance and predisposes to adverse left ventricular remodeling after pressure overload
.
Am J Physiol Heart Circ Physiol
2008
;
295
:
H2495
H2502
.

12

Reddy
SS
,
Agarwal
H
,
Barthwal
MK
.
Cilostazol ameliorates heart failure with preserved ejection fraction and diastolic dysfunction in obese and non-obese hypertensive mice
.
J Mol Cell Cardiol
2018
;
123
:
46
57
.

13

Shevalye
H
,
Lupachyk
S
,
Watcho
P
,
Stavniichuk
R
,
Khazim
K
,
Abboud
HE
,
Obrosova
IG
.
Prediabetic nephropathy as an early consequence of the high-calorie/high-fat diet: relation to oxidative stress
.
Endocrinology
2012
;
153
:
1152
1161
.

14

Dissard
R
,
Klein
J
,
Caubet
C
,
Breuil
B
,
Siwy
J
,
Hoffman
J
,
Sicard
L
,
Ducasse
L
,
Rascalou
S
,
Payre
B
,
Buleon
M
,
Mullen
W
,
Mischak
H
,
Tack
I
,
Bascands
JL
,
Buffin-Meyer
B
,
Schanstra
JP
.
Long term metabolic syndrome induced by a high fat high fructose diet leads to minimal renal injury in C57BL/6 mice
.
PLoS One
2013
;
8
:
e76703
.

15

Christoffersen
C
,
Bollano
E
,
Lindegaard
ML
,
Bartels
ED
,
Goetze
JP
,
Andersen
CB
,
Nielsen
LB
.
Cardiac lipid accumulation associated with diastolic dysfunction in obese mice
.
Endocrinology
2003
;
144
:
3483
3490
.

16

Dong
F
,
Zhang
X
,
Yang
X
,
Esberg
LB
,
Yang
H
,
Zhang
Z
,
Culver
B
,
Ren
J
.
Impaired cardiac contractile function in ventricular myocytes from leptin-deficient ob/ob obese mice
.
J Endocrinol
2006
;
188
:
25
36
.

17

Lindstrom
P
.
The physiology of obese-hyperglycemic mice [ob/ob mice]
.
ScientificWorldJournal
2007
;
7
:
666
685
.

18

Schiattarella
GG
,
Altamirano
F
,
Tong
D
,
French
KM
,
Villalobos
E
,
Kim
SY
,
Luo
X
,
Jiang
N
,
May
HI
,
Wang
ZV
,
Hill
TM
,
Mammen
PPA
,
Huang
J
,
Lee
DI
,
Hahn
VS
,
Sharma
K
,
Kass
DA
,
Lavandero
S
,
Gillette
TG
,
Hill
JA
.
Nitrosative stress drives heart failure with preserved ejection fraction
.
Nature
2019
;
568
:
351
356
.

19

Chiu
HC
,
Kovacs
A
,
Ford
DA
,
Hsu
FF
,
Garcia
R
,
Herrero
P
,
Saffitz
JE
,
Schaffer
JE
.
A novel mouse model of lipotoxic cardiomyopathy
.
J Clin Invest
2001
;
107
:
813
822
.

20

Schilling
JD
,
Machkovech
HM
,
Kim
AH
,
Schwendener
R
,
Schaffer
JE
.
Macrophages modulate cardiac function in lipotoxic cardiomyopathy
.
Am J Physiol Heart Circ Physiol
2012
;
303
:
H1366
H1373
.

21

Yagyu
H
,
Chen
G
,
Yokoyama
M
,
Hirata
K
,
Augustus
A
,
Kako
Y
,
Seo
T
,
Hu
Y
,
Lutz
EP
,
Merkel
M
,
Bensadoun
A
,
Homma
S
,
Goldberg
IJ
.
Lipoprotein lipase (LpL) on the surface of cardiomyocytes increases lipid uptake and produces a cardiomyopathy
.
J Clin Invest
2003
;
111
:
419
426
.

22

Finck
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
.

23

Chiu
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
.

24

Flagg
TP
,
Cazorla
O
,
Remedi
MS
,
Haim
TE
,
Tones
MA
,
Bahinski
A
,
Numann
RE
,
Kovacs
A
,
Schaffer
JE
,
Nichols
CG
,
Nerbonne
JM
.
Ca2+-independent alterations in diastolic sarcomere length and relaxation kinetics in a mouse model of lipotoxic diabetic cardiomyopathy
.
Circ Res
2009
;
104
:
95
103
.

25

Schoiswohl
G
,
Schweiger
M
,
Schreiber
R
,
Gorkiewicz
G
,
Preiss-Landl
K
,
Taschler
U
,
Zierler
KA
,
Radner
FP
,
Eichmann
TO
,
Kienesberger
PC
,
Eder
S
,
Lass
A
,
Haemmerle
G
,
Alsted
TJ
,
Kiens
B
,
Hoefler
G
,
Zechner
R
,
Zimmermann
R
.
Adipose triglyceride lipase plays a key role in the supply of the working muscle with fatty acids
.
J Lipid Res
2010
;
51
:
490
499
.

26

Wang
X
,
McLennan
SV
,
Allen
TJ
,
Tsoutsman
T
,
Semsarian
C
,
Twigg
SM
.
Adverse effects of high glucose and free fatty acid on cardiomyocytes are mediated by connective tissue growth factor
.
Am J Physiol Cell Physiol
2009
;
297
:
C1490
C1500
.

27

Wang
Y
,
Ebermann
L
,
Sterner-Kock
A
,
Wika
S
,
Schultheiss
HP
,
Dorner
A
,
Walther
T
.
Myocardial overexpression of adenine nucleotide translocase 1 ameliorates diabetic cardiomyopathy in mice
.
Exp Physiol
2009
;
94
:
220
227
.

28

Huynh
K
,
McMullen
JR
,
Julius
TL
,
Tan
JW
,
Love
JE
,
Cemerlang
N
,
Kiriazis
H
,
Du
XJ
,
Ritchie
RH
.
Cardiac-specific IGF-1 receptor transgenic expression protects against cardiac fibrosis and diastolic dysfunction in a mouse model of diabetic cardiomyopathy
.
Diabetes
2010
;
59
:
1512
1520
.

29

Yu
X
,
Tesiram
YA
,
Towner
RA
,
Abbott
A
,
Patterson
E
,
Huang
S
,
Garrett
MW
,
Chandrasekaran
S
,
Matsuzaki
S
,
Szweda
LI
,
Gordon
BE
,
Kem
DC
.
Early myocardial dysfunction in streptozotocin-induced diabetic mice: a study using in vivo magnetic resonance imaging (MRI)
.
Cardiovasc Diabetol
2007
;
6
:
6
.

30

Trost
SU
,
Belke
DD
,
Bluhm
WF
,
Meyer
M
,
Swanson
E
,
Dillmann
WH
.
Overexpression of the sarcoplasmic reticulum ca(2+)-ATPase improves myocardial contractility in diabetic cardiomyopathy
.
Diabetes
2002
;
51
:
1166
1171
.

31

Wang
J
,
Song
Y
,
Elsherif
L
,
Song
Z
,
Zhou
G
,
Prabhu
SD
,
Saari
JT
,
Cai
L
.
Cardiac metallothionein induction plays the major role in the prevention of diabetic cardiomyopathy by zinc supplementation
.
Circulation
2006
;
113
:
544
554
.

32

Srinivasan
K
,
Ramarao
P
.
Animal models in type 2 diabetes research: an overview
.
Indian J Med Res
2007
;
125
:
451
472
.

33

Semeniuk
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
H982
.

34

Jiang
T
,
Wang
XX
,
Scherzer
P
,
Wilson
P
,
Tallman
J
,
Takahashi
H
,
Li
J
,
Iwahashi
M
,
Sutherland
E
,
Arend
L
,
Levi
M
.
Farnesoid X receptor modulates renal lipid metabolism, fibrosis, and diabetic nephropathy
.
Diabetes
2007
;
56
:
2485
2493
.

35

Sharma
K
,
McCue
P
,
Dunn
SR
.
Diabetic kidney disease in the db/db mouse
.
Am J Physiol Renal Physiol
2003
;
284
:
F1138
F1144
.

36

Gui
T
,
Li
Y
,
Zhang
S
,
Zhang
N
,
Sun
Y
,
Liu
F
,
Chen
Q
,
Gai
Z
.
Docosahexaenoic acid protects against palmitate-induced mitochondrial dysfunction in diabetic cardiomyopathy
.
Biomed Pharmacother
2020
;
128
:
110306
.

37

Han
Q
,
Yeung
SC
,
Ip
MSM
,
Mak
JCW
.
Dysregulation of cardiac lipid parameters in high-fat high-cholesterol diet-induced rat model
.
Lipids Health Dis
2018
;
17
:
255
.

38

Ruiz-Hurtado
G
,
Garcia-Prieto
CF
,
Pulido-Olmo
H
,
Velasco-Martin
JP
,
Villa-Valverde
P
,
Fernandez-Valle
ME
,
Bosca
L
,
Fernandez-Velasco
M
,
Regadera
J
,
Somoza
B
,
Fernandez-Alfonso
MS
.
Mild and short-term caloric restriction prevents obesity-induced cardiomyopathy in young Zucker rats without changing in metabolites and fatty acids cardiac profile
.
Front Physiol
2017
;
8
:
42
.

39

Moran
TH
.
Unraveling the obesity of OLETF rats
.
Physiol Behav
2008
;
94
:
71
78
.

40

Murase
T
,
Hattori
T
,
Ohtake
M
,
Abe
M
,
Amakusa
Y
,
Takatsu
M
,
Murohara
T
,
Nagata
K
.
Cardiac remodeling and diastolic dysfunction in DahlS.Z-lepr(fa)/lepr(fa) rats: a new animal model of metabolic syndrome
.
Hypertens Res
2012
;
35
:
186
193
.

41

Loganathan
R
,
Bilgen
M
,
Al-Hafez
B
,
Alenezy
MD
,
Smirnova
IV
.
Cardiac dysfunction in the diabetic rat: quantitative evaluation using high resolution magnetic resonance imaging
.
Cardiovasc Diabetol
2006
;
5
:
7
.

42

Hamblin
M
,
Friedman
DB
,
Hill
S
,
Caprioli
RM
,
Smith
HM
,
Hill
MF
.
Alterations in the diabetic myocardial proteome coupled with increased myocardial oxidative stress underlies diabetic cardiomyopathy
.
J Mol Cell Cardiol
2007
;
42
:
884
895
.

43

Sun
D
,
Shen
M
,
Li
J
,
Li
W
,
Zhang
Y
,
Zhao
L
,
Zhang
Z
,
Yuan
Y
,
Wang
H
,
Cao
F
.
Cardioprotective effects of tanshinone IIA pretreatment via kinin B2 receptor-akt-GSK-3beta dependent pathway in experimental diabetic cardiomyopathy
.
Cardiovasc Diabetol
2011
;
10
:
4
.

44

Tschope
C
,
Walther
T
,
Koniger
J
,
Spillmann
F
,
Westermann
D
,
Escher
F
,
Pauschinger
M
,
Pesquero
JB
,
Bader
M
,
Schultheiss
HP
,
Noutsias
M
.
Prevention of cardiac fibrosis and left ventricular dysfunction in diabetic cardiomyopathy in rats by transgenic expression of the human tissue kallikrein gene
.
FASEB J
2004
;
18
:
828
835
.

45

Lin
G
,
Craig
GP
,
Zhang
L
,
Yuen
VG
,
Allard
M
,
McNeill
JH
,
MacLeod
KM
.
Acute inhibition of rho-kinase improves cardiac contractile function in streptozotocin-diabetic rats
.
Cardiovasc Res
2007
;
75
:
51
58
.

46

Wichi
R
,
Malfitano
C
,
Rosa
K
,
De Souza
SB
,
Salemi
V
,
Mostarda
C
,
De Angelis
K
,
Irigoyen
MC
.
Noninvasive and invasive evaluation of cardiac dysfunction in experimental diabetes in rodents
.
Cardiovasc Diabetol
2007
;
6
:
14
.

47

van den Brom
CE
,
Bosmans
JW
,
Vlasblom
R
,
Handoko
LM
,
Huisman
MC
,
Lubberink
M
,
Molthoff
CF
,
Lammertsma
AA
,
Ouwens
MD
,
Diamant
M
,
Boer
C
.
Diabetic cardiomyopathy in Zucker diabetic fatty rats: the forgotten right ventricle
.
Cardiovasc Diabetol
2010
;
9
:
25
.

48

Forcheron
F
,
Basset
A
,
Abdallah
P
,
Del Carmine
P
,
Gadot
N
,
Beylot
M
.
Diabetic cardiomyopathy: effects of fenofibrate and metformin in an experimental model–the Zucker diabetic rat
.
Cardiovasc Diabetol
2009
;
8
:
16
.

49

Allwood
MA
,
Foster
AJ
,
Arkell
AM
,
Beaudoin
MS
,
Snook
LA
,
Romanova
N
,
Murrant
CL
,
Holloway
GP
,
Wright
DC
,
Simpson
JA
.
Respiratory muscle weakness in the Zucker diabetic fatty rat
.
Am J Physiol Regul Integr Comp Physiol
2015
;
309
:
R780
R787
.

50

Kasiske
BL
,
O'Donnell
MP
,
Cleary
MP
,
Keane
WF
.
Treatment of hyperlipidemia reduces glomerular injury in obese Zucker rats
.
Kidney Int
1988
;
33
:
667
672
.

51

Shimoshige
Y
,
Ikuma
K
,
Yamamoto
T
,
Takakura
S
,
Kawamura
I
,
Seki
J
,
Mutoh
S
,
Goto
T
.
The effects of zenarestat, an aldose reductase inhibitor, on peripheral neuropathy in Zucker diabetic fatty rats
.
Metab Clin Exp
2000
;
49
:
1395
1399
.

52

Schmidt
RE
,
Dorsey
DA
,
Beaudet
LN
,
Peterson
RG
.
Analysis of the Zucker diabetic fatty (ZDF) type 2 diabetic rat model suggests a neurotrophic role for insulin/IGF-I in diabetic autonomic neuropathy
.
Am J Pathol
2003
;
163
:
21
28
.

53

Shang
J
,
Chen
Z
,
Wang
M
,
Li
Q
,
Feng
W
,
Wu
Y
,
Wu
W
,
Graziano
MP
,
Chintala
M
.
Zucker diabetic fatty rats exhibit hypercoagulability and accelerated thrombus formation in the arterio-venous shunt model of thrombosis
.
Thromb Res
2014
;
134
:
433
439
.

54

Gronholm
T
,
Cheng
ZJ
,
Palojoki
E
,
Eriksson
A
,
Backlund
T
,
Vuolteenaho
O
,
Finckenberg
P
,
Laine
M
,
Mervaala
E
,
Tikkanen
I
.
Vasopeptidase inhibition has beneficial cardiac effects in spontaneously diabetic Goto-Kakizaki rats
.
Eur J Pharmacol
2005
;
519
:
267
276
.

55

D’Souza
A
,
Howarth
FC
,
Yanni
J
,
Dobryznski
H
,
Boyett
MR
,
Adeghate
E
,
Bidasee
KR
,
Singh
J
.
Left ventricle structural remodelling in the prediabetic Goto-Kakizaki rat
.
Exp Physiol
2011
;
96
:
875
888
.

56

Karahashi
M
,
Hirata-Hanta
Y
,
Kawabata
K
,
Tsutsumi
D
,
Kametani
M
,
Takamatsu
N
,
Sakamoto
T
,
Yamazaki
T
,
Asano
S
,
Mitsumoto
A
,
Kawashima
Y
,
Kudo
N
.
Abnormalities in the metabolism of fatty acids and triacylglycerols in the liver of the Goto-Kakizaki rat: a model for non-obese type 2 diabetes
.
Lipids
2016
;
51
:
955
971
.

57

Meagher
P
,
Civitarese
R
,
Lee
X
,
Gordon
M
,
Bugyei-Twum
A
,
Desjardins
JF
,
Kabir
G
,
Zhang
Y
,
Kosanam
H
,
Visram
A
,
Leong-Poi
H
,
Advani
A
,
Connelly
KA
.
The Goto Kakizaki rat: impact of age upon changes in cardiac and renal structure, function
.
PLoS One
2021
;
16
:
e0252711
.

58

Masiello
P
,
Broca
C
,
Gross
R
,
Roye
M
,
Manteghetti
M
,
Hillaire-Buys
D
,
Novelli
M
,
Ribes
G
.
Experimental NIDDM: development of a new model in adult rats administered streptozotocin and nicotinamide
.
Diabetes
1998
;
47
:
224
229
.

59

Ding
G
,
Li
L
,
Zhang
L
,
Chopp
M
,
Davoodi-Bojd
E
,
Li
Q
,
Li
C
,
Wei
M
,
Zhang
Z
,
Jiang
Q
.
MRI Metrics of cerebral endothelial cell-derived exosomes for the treatment of cognitive dysfunction induced in aging rats subjected to type 2 diabetes
.
Diabetes
2022
;
71
:
873
880
.

60

Olaniyi
KS
,
Amusa
OA
.
Sodium acetate-mediated inhibition of histone deacetylase alleviates hepatic lipid dysregulation and its accompanied injury in streptozotocin-nicotinamide-induced diabetic rats
.
Biomed Pharmacother
2020
;
128
:
110226
.

61

Jesmin
S
,
Shima
T
,
Soya
M
,
Takahashi
K
,
Omura
K
,
Ogura
K
,
Koizumi
H
,
Soya
H
.
Long-term light and moderate exercise intervention similarly prevent both hippocampal and glycemic dysfunction in presymptomatic type 2 diabetic rats
.
Am J Physiol Endocrinol Metab
2022
;
322
:
E219
E230
.

62

Kawano
K
,
Hirashima
T
,
Mori
S
,
Natori
T
.
OLETF (Otsuka long-evans tokushima fatty) rat: a new NIDDM rat strain
.
Diabetes Res Clin Pract
1994
;
24
:
S317
S320
.

63

Du
J
,
Zhu
M
,
Li
H
,
Liang
G
,
Li
Y
,
Feng
S
.
Metformin attenuates cardiac remodeling in mice through the Nrf2/Keap1 signaling pathway
.
Exp Ther Med
2020
;
20
:
838
845
.

64

Wu
L
,
Wang
K
,
Wang
W
,
Wen
Z
,
Wang
P
,
Liu
L
,
Wang
DW
.
Glucagon-like peptide-1 ameliorates cardiac lipotoxicity in diabetic cardiomyopathy via the PPARalpha pathway
.
Aging Cell
2018
;
17
:
e12763
.

65

McMurray
JJV
,
Ponikowski
P
,
Bolli
GB
,
Lukashevich
V
,
Kozlovski
P
,
Kothny
W
,
Lewsey
JD
,
Krum
H
,
VIVIDD Trial Committees and Investigators
.
Effects of vildagliptin on ventricular function in patients with type 2 diabetes mellitus and heart failure: a randomized placebo-controlled trial
.
JACC Heart Fail
2018
;
6
:
8
17
.

66

Xu
Y
,
Wang
T
,
Yang
Z
,
Lin
H
,
Shen
P
,
Zhan
S
.
Sulphonylureas monotherapy and risk of hospitalization for heart failure in patients with type 2 diabetes mellitus: a population-based cohort study in China
.
Pharmacoepidemiol Drug Saf
2020
;
29
:
635
643
.

67

Pocock
SJ
,
Wang
D
,
Pfeffer
MA
,
Yusuf
S
,
McMurray
JJ
,
Swedberg
KB
,
Ostergren
J
,
Michelson
EL
,
Pieper
KS
,
Granger
CB
.
Predictors of mortality and morbidity in patients with chronic heart failure
.
Eur Heart J
2006
;
27
:
65
75
.

68

Lago
RM
,
Singh
PP
,
Nesto
RW
.
Congestive heart failure and cardiovascular death in patients with prediabetes and type 2 diabetes given thiazolidinediones: a meta-analysis of randomised clinical trials
.
Lancet
2007
;
370
:
1129
1136
.

69

Scirica
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
.

70

Clee
SM
,
Attie
AD
.
The genetic landscape of type 2 diabetes in mice
.
Endocr Rev
2007
;
28
:
48
83
.

71

Ishikawa
M
,
Saito
K
,
Urata
M
,
Kumagai
Y
,
Maekawa
K
,
Saito
Y
.
Comparison of circulating lipid profiles between fasting humans and three animal species used in preclinical studies: mice, rats and rabbits
.
Lipids Health Dis
2015
;
14
:
104
.

72

Nanayakkara
N
,
Curtis
AJ
,
Heritier
S
,
Gadowski
AM
,
Pavkov
ME
,
Kenealy
T
,
Owens
DR
,
Thomas
RL
,
Song
S
,
Wong
J
,
Chan
JC
,
Luk
AO
,
Penno
G
,
Ji
L
,
Mohan
V
,
Amutha
A
,
Romero-Aroca
P
,
Gasevic
D
,
Magliano
DJ
,
Teede
HJ
,
Chalmers
J
,
Zoungas
S
.
Impact of age at type 2 diabetes mellitus diagnosis on mortality and vascular complications: systematic review and meta-analyses
.
Diabetologia
2021
;
64
:
275
287
.

73

Song
SH
,
Hardisty
CA
.
Early-onset type 2 diabetes mellitus: an increasing phenomenon of elevated cardiovascular risk
.
Expert Rev Cardiovasc Ther
2008
;
6
:
315
322
.

74

Poornima
IG
,
Parikh
P
,
Shannon
RP
.
Diabetic cardiomyopathy: the search for a unifying hypothesis
.
Circ Res
2006
;
98
:
596
605
.

75

Tan
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
.

76

Bertero
E
,
Maack
C
.
Calcium signaling and reactive oxygen species in mitochondria
.
Circ Res
2018
;
122
:
1460
1478
.

77

Xu
J
,
Zhou
Q
,
Xu
W
,
Cai
L
.
Endoplasmic reticulum stress and diabetic cardiomyopathy
.
Exp Diabetes Res
2012
;
2012
:
827971
.

78

Dia
M
,
Gomez
L
,
Thibault
H
,
Tessier
N
,
Leon
C
,
Chouabe
C
,
Ducreux
S
,
Gallo-Bona
N
,
Tubbs
E
,
Bendridi
N
,
Chanon
S
,
Leray
A
,
Belmudes
L
,
Coute
Y
,
Kurdi
M
,
Ovize
M
,
Rieusset
J
,
Paillard
M
.
Reduced reticulum-mitochondria ca(2+) transfer is an early and reversible trigger of mitochondrial dysfunctions in diabetic cardiomyopathy
.
Basic Res Cardiol
2020
;
115
:
74
.

79

Nirengi
S
,
Peres Valgas da Silva
C
,
Stanford
KI
.
Disruption of energy utilization in diabetic cardiomyopathy; a mini review
.
Curr Opin Pharmacol
2020
;
54
:
82
90
.

80

Kaludercic
N
,
Di Lisa
F
.
Mitochondrial ROS formation in the pathogenesis of diabetic cardiomyopathy
.
Front Cardiovasc Med
2020
;
7
:
12
.

81

Westermeier
F
,
Navarro-Marquez
M
,
Lopez-Crisosto
C
,
Bravo-Sagua
R
,
Quiroga
C
,
Bustamante
M
,
Verdejo
HE
,
Zalaquett
R
,
Ibacache
M
,
Parra
V
,
Castro
PF
,
Rothermel
BA
,
Hill
JA
,
Lavandero
S
.
Defective insulin signaling and mitochondrial dynamics in diabetic cardiomyopathy
.
Biochim Biophys Acta
2015
;
1853
:
1113
1118
.

82

Tong
M
,
Saito
T
,
Zhai
P
,
Oka
SI
,
Mizushima
W
,
Nakamura
M
,
Ikeda
S
,
Shirakabe
A
,
Sadoshima
J
.
Mitophagy is essential for maintaining cardiac function during high fat diet-induced diabetic cardiomyopathy
.
Circ Res
2019
;
124
:
1360
1371
.

83

Wu
S
,
Lu
Q
,
Ding
Y
,
Wu
Y
,
Qiu
Y
,
Wang
P
,
Mao
X
,
Huang
K
,
Xie
Z
,
Zou
MH
.
Hyperglycemia-driven inhibition of AMP-activated protein kinase alpha2 induces diabetic cardiomyopathy by promoting mitochondria-associated endoplasmic reticulum membranes in vivo
.
Circulation
2019
;
139
:
1913
1936
.

84

Riehle
C
,
Bauersachs
J
.
Of mice and men: models and mechanisms of diabetic cardiomyopathy
.
Basic Res Cardiol
2018
;
114
:
2
.

85

Marwick
TH
,
Gimelli
A
,
Plein
S
,
Bax
JJ
,
Charron
P
,
Delgado
V
,
Donal
E
,
Lancellotti
P
,
Levelt
E
,
Maurovich-Horvat
P
,
Neubauer
S
,
Pontone
G
,
Saraste
A
,
Cosyns
B
,
Edvardsen
T
,
Popescu
BA
,
Galderisi
M
,
Derumeaux
G
.
Reviewers: This document was reviewed by members of the ESDC
,
Back
M
,
Bertrand
PB
,
Dweck
M
,
Keenan
N
,
Magne
J
,
Neglia
D
,
Stankovic
I
.
Multimodality imaging approach to left ventricular dysfunction in diabetes: an expert consensus document from the European association of cardiovascular imaging
.
Eur Heart J Cardiovasc Imaging
2021
, 23(2):e62-e84.

86

Czibik
G
,
Mezdari
Z
,
Murat Altintas
D
,
Brehat
J
,
Pini
M
,
d'Humieres
T
,
Delmont
T
,
Radu
C
,
Breau
M
,
Liang
H
,
Martel
C
,
Abatan
A
,
Sarwar
R
,
Marion
O
,
Naushad
S
,
Zhang
Y
,
Halfaoui
M
,
Suffee
N
,
Morin
D
,
Adnot
S
,
Hatem
S
,
Yavari
A
,
Sawaki
D
,
Derumeaux
G
.
Dysregulated phenylalanine catabolism plays a key role in the trajectory of cardiac aging
.
Circulation
2021
;
144
:
559
574
.

87

Palmer
AK
,
Gustafson
B
,
Kirkland
JL
,
Smith
U
.
Cellular senescence: at the nexus between ageing and diabetes
.
Diabetologia
2019
;
62
:
1835
1841
.

88

Cianflone
E
,
Torella
M
,
Biamonte
F
,
De Angelis
A
,
Urbanek
K
,
Costanzo
FS
,
Rota
M
,
Ellison-Hughes
GM
,
Torella
D
.
Targeting cardiac stem cell senescence to treat cardiac aging and disease
.
Cells
2020
;
9
(6):1558.

89

Anderson
R
,
Lagnado
A
,
Maggiorani
D
,
Walaszczyk
A
,
Dookun
E
,
Chapman
J
,
Birch
J
,
Salmonowicz
H
,
Ogrodnik
M
,
Jurk
D
,
Proctor
C
,
Correia-Melo
C
,
Victorelli
S
,
Fielder
E
,
Berlinguer-Palmini
R
,
Owens
A
,
Greaves
LC
,
Kolsky
KL
,
Parini
A
,
Douin-Echinard
V
,
LeBrasseur
NK
,
Arthur
HM
,
Tual-Chalot
S
,
Schafer
MJ
,
Roos
CM
,
Miller
JD
,
Robertson
N
,
Mann
J
,
Adams
PD
,
Tchkonia
T
,
Kirkland
JL
,
Mialet-Perez
J
,
Richardson
GD
,
Passos
JF
.
Length-independent telomere damage drives post-mitotic cardiomyocyte senescence
.
EMBO J
2019
;
38
(5):e100492.

90

Costantino
S
,
Paneni
F
,
Luscher
TF
,
Cosentino
F
.
MicroRNA profiling unveils hyperglycaemic memory in the diabetic heart
.
Eur Heart J
2016
;
37
:
572
576
.

91

Withaar
C
,
Meems
LMG
,
Markousis-Mavrogenis
G
,
Boogerd
CJ
,
Sillje
HHW
,
Schouten
EM
,
Dokter
MM
,
Voors
AA
,
Westenbrink
BD
,
Lam
CSP
,
de Boer
RA
.
The effects of liraglutide and dapagliflozin on cardiac function and structure in a multi-hit mouse model of heart failure with preserved ejection fraction
.
Cardiovasc Res
2021
;
117
:
2108
2124
.

92

Palmer
AK
,
Xu
M
,
Zhu
Y
,
Pirtskhalava
T
,
Weivoda
MM
,
Hachfeld
CM
,
Prata
LG
,
van Dijk
TH
,
Verkade
E
,
Casaclang-Verzosa
G
,
Johnson
KO
,
Cubro
H
,
Doornebal
EJ
,
Ogrodnik
M
,
Jurk
D
,
Jensen
MD
,
Chini
EN
,
Miller
JD
,
Matveyenko
A
,
Stout
MB
,
Schafer
MJ
,
White
TA
,
Hickson
LJ
,
Demaria
M
,
Garovic
V
,
Grande
J
,
Arriaga
EA
,
Kuipers
F
,
von Zglinicki
T
,
LeBrasseur
NK
,
Campisi
J
,
Tchkonia
T
,
Kirkland
JL
.
Targeting senescent cells alleviates obesity-induced metabolic dysfunction
.
Aging Cell
2019
;
18
:
e12950
.

93

Palmer
AK
,
Tchkonia
T
,
LeBrasseur
NK
,
Chini
EN
,
Xu
M
,
Kirkland
JL
.
Cellular senescence in type 2 diabetes: A therapeutic opportunity
.
Diabetes
2015
;
64
:
2289
2298
.

94

Ernande
L
,
Audureau
E
,
Jellis
CL
,
Bergerot
C
,
Henegar
C
,
Sawaki
D
,
Czibik
G
,
Volpi
C
,
Canoui-Poitrine
F
,
Thibault
H
,
Ternacle
J
,
Moulin
P
,
Marwick
TH
,
Derumeaux
G
.
Clinical implications of echocardiographic phenotypes of patients with diabetes mellitus
.
J Am Coll Cardiol
2017
;
70
:
1704
1716
.

95

Galderisi
M
,
Anderson
KM
,
Wilson
PW
,
Levy
D
.
Echocardiographic evidence for the existence of a distinct diabetic cardiomyopathy (the framingham heart study)
.
Am J Cardiol
1991
;
68
:
85
89
.

96

Lum-Naihe
K
,
Toedebusch
R
,
Mahmood
A
,
Bajwa
J
,
Carmack
T
,
Kumar
SA
,
Ardhanari
S
,
DeMarco
VG
,
Emter
CA
,
Pulakat
L
.
Cardiovascular disease progression in female Zucker diabetic fatty rats occurs via unique mechanisms compared to males
.
Sci Rep
2017
;
7
:
17823
.

97

Desrois
M
,
Sidell
RJ
,
Gauguier
D
,
Davey
CL
,
Radda
GK
,
Clarke
K
.
Gender differences in hypertrophy, insulin resistance and ischemic injury in the aging type 2 diabetic rat heart
.
J Mol Cell Cardiol
2004
;
37
:
547
555
.

98

Bowden
MA
,
Tesch
GH
,
Julius
TL
,
Rosli
S
,
Love
JE
,
Ritchie
RH
.
Earlier onset of diabesity-induced adverse cardiac remodeling in female compared to male mice
.
Obesity
2015
;
23
:
1166
1177
.

99

Toedebusch
R
,
Belenchia
A
,
Pulakat
L
.
Diabetic cardiomyopathy: impact of biological sex on disease development and molecular signatures
.
Front Physiol
2018
;
9
:
453
.

100

Ventura-Clapier
R
,
Dworatzek
E
,
Seeland
U
,
Kararigas
G
,
Arnal
JF
,
Brunelleschi
S
,
Carpenter
TC
,
Erdmann
J
,
Franconi
F
,
Giannetta
E
,
Glezerman
M
,
Hofmann
SM
,
Junien
C
,
Katai
M
,
Kublickiene
K
,
Konig
IR
,
Majdic
G
,
Malorni
W
,
Mieth
C
,
Miller
VM
,
Reynolds
RM
,
Shimokawa
H
,
Tannenbaum
C
,
D’Ursi
AM
,
Regitz-Zagrosek
V
.
Sex in basic research: concepts in the cardiovascular field
.
Cardiovasc Res
2017
;
113
:
711
724
.

101

Tramunt
B
,
Smati
S
,
Grandgeorge
N
,
Lenfant
F
,
Arnal
JF
,
Montagner
A
,
Gourdy
P
.
Sex differences in metabolic regulation and diabetes susceptibility
.
Diabetologia
2020
;
63
:
453
461
.

102

Leskanicova
A
,
Chovancova
O
,
Babincak
M
,
Verboova
L
,
Benetinova
Z
,
Macekova
D
,
Kostolny
J
,
Smajda
B
,
Kiskova
T
.
Sexual dimorphism in energy metabolism of Wistar rats using data analysis
.
Molecules
2020
;
25
(10):2353.

103

Reichelt
ME
,
Mellor
KM
,
Bell
JR
,
Chandramouli
C
,
Headrick
JP
,
Delbridge
LM
.
Sex, sex steroids, and diabetic cardiomyopathy: making the case for experimental focus
.
Am J Physiol Heart Circ Physiol
2013
;
305
:
H779
H792
.

104

Devanathan
S
,
Whitehead
TD
,
Fettig
N
,
Gropler
RJ
,
Nemanich
S
,
Shoghi
KI
.
Sexual dimorphism in myocardial acylcarnitine and triglyceride metabolism
.
Biol Sex Differ
2016
;
7
:
25
.

105

Lu
HS
,
Schmidt
AM
,
Hegele
RA
,
Mackman
N
,
Rader
DJ
,
Weber
C
,
Daugherty
A
.
Reporting sex and sex differences in preclinical studies
.
Arterioscler Thromb Vasc Biol
2018
;
38
:
e171
e184
.

106

Perrone-Filardi
P
,
Paolillo
S
,
Costanzo
P
,
Savarese
G
,
Trimarco
B
,
Bonow
RO
.
The role of metabolic syndrome in heart failure
.
Eur Heart J
2015
;
36
:
2630
2634
.

107

Alpert
MA
.
Obesity cardiomyopathy: pathophysiology and evolution of the clinical syndrome
.
Am J Med Sci
2001
;
321
:
225
236
.

108

Kenchaiah
S
,
Evans
JC
,
Levy
D
,
Wilson
PW
,
Benjamin
EJ
,
Larson
MG
,
Kannel
WB
,
Vasan
RS
.
Obesity and the risk of heart failure
.
N Engl J Med
2002
;
347
:
305
313
.

109

Rayner
JJ
,
Peterzan
MA
,
Clarke
WT
,
Rodgers
CT
,
Neubauer
S
,
Rider
OJ
.
Obesity modifies the energetic phenotype of dilated cardiomyopathy
.
Eur Heart J
2021
;43(9):868–77.

110

Heydemann
A
.
An overview of murine high fat diet as a model for type 2 diabetes mellitus
.
J Diabetes Res
.
2016
;
2016
:
2902351
.

111

Abbasi
SA
,
Hundley
WG
,
Bluemke
DA
,
Jerosch-Herold
M
,
Blankstein
R
,
Petersen
SE
,
Rider
OJ
,
Lima
JA
,
Allison
MA
,
Murthy
VL
,
Shah
RV
.
Visceral adiposity and left ventricular remodeling: the multi-ethnic study of atherosclerosis
.
Nutr Metab Cardiovasc Dis
2015
;
25
:
667
676
.

112

Arderiu
G
,
Lambert
C
,
Ballesta
C
,
Moscatiello
F
,
Vilahur
G
,
Badimon
L
.
Cardiovascular risk factors and differential transcriptomic profile of the subcutaneous and visceral adipose tissue and their resident stem cells
.
Cells
2020
;
9
.

113

Sawaki
D
,
Czibik
G
,
Pini
M
,
Ternacle
J
,
Suffee
N
,
Mercedes
R
,
Marcelin
G
,
Surenaud
M
,
Marcos
E
,
Gual
P
,
Clement
K
,
Hue
S
,
Adnot
S
,
Hatem
SN
,
Tsuchimochi
I
,
Yoshimitsu
T
,
Henegar
C
,
Derumeaux
G
.
Visceral adipose tissue drives cardiac aging through modulation of fibroblast senescence by osteopontin production
.
Circulation
2018
;
138
:
809
822
.

114

Venteclef
N
,
Guglielmi
V
,
Balse
E
,
Gaborit
B
,
Cotillard
A
,
Atassi
F
,
Amour
J
,
Leprince
P
,
Dutour
A
,
Clement
K
,
Hatem
SN
.
Human epicardial adipose tissue induces fibrosis of the atrial myocardium through the secretion of adipo-fibrokines
.
Eur Heart J
2015
;
36
:
795
805a
.

115

Liakopoulos
V
,
Franzen
S
,
Svensson
AM
,
Sattar
N
,
Miftaraj
M
,
Bjorck
S
,
Ottosson
J
,
Naslund
I
,
Gudbjornsdottir
S
,
Eliasson
B
.
Renal and cardiovascular outcomes after weight loss from gastric bypass surgery in type 2 diabetes: cardiorenal risk reductions exceed atherosclerotic benefits
.
Diabetes Care
2020
;
43
:
1276
1284
.

116

Koshino
Y
,
Villarraga
HR
,
Somers
VK
,
Miranda
WR
,
Garza
CA
,
Hsiao
JF
,
Yu
Y
,
Saleh
HK
,
Lopez-Jimenez
F
.
Changes in myocardial mechanics in patients with obesity following major weight loss after bariatric surgery
.
Obesity
2013
;
21
:
1111
1118
.

117

Pelliccia
A
,
Sharma
S
,
Gati
S
,
Back
M
,
Borjesson
M
,
Caselli
S
,
Collet
JP
,
Corrado
D
,
Drezner
JA
,
Halle
M
,
Hansen
D
,
Heidbuchel
H
,
Myers
J
,
Niebauer
J
,
Papadakis
M
,
Piepoli
MF
,
Prescott
E
,
Roos-Hesselink
JW
,
Graham Stuart
A
,
Taylor
RS
,
Thompson
PD
,
Tiberi
M
,
Vanhees
L
,
Wilhelm
M
,
ESC Scientific Document Group
.
2020 ESC guidelines on sports cardiology and exercise in patients with cardiovascular disease
.
Eur Heart J
2020
;
42
:
17
96
.

118

Veeranki
S
,
Givvimani
S
,
Kundu
S
,
Metreveli
N
,
Pushpakumar
S
,
Tyagi
SC
.
Moderate intensity exercise prevents diabetic cardiomyopathy associated contractile dysfunction through restoration of mitochondrial function and connexin 43 levels in db/db mice
.
J Mol Cell Cardiol
2016
;
92
:
163
173
.

119

Pini
M
,
Czibik
G
,
Sawaki
D
,
Mezdari
Z
,
Braud
L
,
Delmont
T
,
Mercedes
R
,
Martel
C
,
Buron
N
,
Marcelin
G
,
Borgne-Sanchez
A
,
Foresti
R
,
Motterlini
R
,
Henegar
C
,
Derumeaux
G
.
Adipose tissue senescence is mediated by increased ATP content after a short-term high-fat diet exposure
.
Aging Cell
2021
;
20
:
e13421
.

120

El-Armouche
A
,
Eschenhagen
T
.
Beta-adrenergic stimulation and myocardial function in the failing heart
.
Heart Fail Rev
2009
;
14
:
225
241
.

121

Pop-Busui
R
.
Cardiac autonomic neuropathy in diabetes: a clinical perspective
.
Diabetes Care
2010
;
33
:
434
441
.

122

Bristow
MR
,
Ginsburg
R
,
Minobe
W
,
Cubicciotti
RS
,
Sageman
WS
,
Lurie
K
,
Billingham
ME
,
Harrison
DC
,
Stinson
EB
.
Decreased catecholamine sensitivity and beta-adrenergic-receptor density in failing human hearts
.
N Engl J Med
1982
;
307
:
205
211
.

123

Ungerer
M
,
Bohm
M
,
Elce
JS
,
Erdmann
E
,
Lohse
MJ
.
Altered expression of beta-adrenergic receptor kinase and beta 1-adrenergic receptors in the failing human heart
.
Circulation
1993
;
87
:
454
463
.

124

Bockus
LB
,
Humphries
KM
.
cAMP-dependent protein kinase (PKA) signaling is impaired in the diabetic heart
.
J Biol Chem
2015
;
290
:
29250
29258
.

125

Metrich
M
,
Lucas
A
,
Gastineau
M
,
Samuel
JL
,
Heymes
C
,
Morel
E
,
Lezoualc’h
F
.
Epac mediates beta-adrenergic receptor-induced cardiomyocyte hypertrophy
.
Circ Res
2008
;
102
:
959
965
.

126

Wang
Q
,
Liu
Y
,
Fu
Q
,
Xu
B
,
Zhang
Y
,
Kim
S
,
Tan
R
,
Barbagallo
F
,
West
T
,
Anderson
E
,
Wei
W
,
Abel
ED
,
Xiang
YK
.
Inhibiting insulin-mediated beta2-adrenergic receptor activation prevents diabetes-associated cardiac dysfunction
.
Circulation
2017
;
135
:
73
88
.

127

Amour
J
,
Loyer
X
,
Le Guen
M
,
Mabrouk
N
,
David
JS
,
Camors
E
,
Carusio
N
,
Vivien
B
,
Andriantsitohaina
R
,
Heymes
C
,
Riou
B
.
Altered contractile response due to increased beta3-adrenoceptor stimulation in diabetic cardiomyopathy: the role of nitric oxide synthase 1-derived nitric oxide
.
Anesthesiology
2007
;
107
:
452
460
.

128

Reutrakul
S
,
Mokhlesi
B
.
Obstructive sleep apnea and diabetes: a state of the art review
.
Chest
2017
;
152
:
1070
1086
.

129

Prabhakar
NR
,
Peng
YJ
,
Nanduri
J
.
Hypoxia-inducible factors and obstructive sleep apnea
.
J Clin Invest
2020
;
130
:
5042
5051
.

130

Wasserman
DH
,
Wang
TJ
,
Brown
NJ
.
The vasculature in prediabetes
.
Circ Res
2018
;
122
:
1135
1150
.

131

Mishra
S
,
Kass
DA
.
Cellular and molecular pathobiology of heart failure with preserved ejection fraction
.
Nat Rev Cardiol
2021
;
18
:
400
423
.

132

Withaar
C
,
Lam
CSP
,
Schiattarella
GG
,
de Boer
RA
,
Meems
LMG
.
Heart failure with preserved ejection fraction in humans and mice: embracing clinical complexity in mouse models
.
Eur Heart J
2021
;
42
:
4420
4430
.

133

Jindal
A
,
Garcia-Touza
M
,
Jindal
N
,
Whaley-Connell
A
,
Sowers
JR
.
Diabetic kidney disease and the cardiorenal syndrome: old disease, new perspectives
.
Endocrinol Metab Clin North Am
2013
;
42
:
789
808
.

134

Palau
V
,
Riera
M
,
Soler
MJ
.
The reno-cardiovascular connection in the patient with diabetes mellitus: what's new?
Endocrinol Diabetes Nutr
2017
;
64
:
237
240
.

135

Ciccarelli
M
,
Dawson
D
,
Falcao-Pires
I
,
Giacca
M
,
Hamdani
N
,
Heymans
S
,
Hooghiemstra
A
,
Leeuwis
A
,
Hermkens
D
,
Tocchetti
CG
,
van der Velden
J
,
Zacchigna
S
,
Thum
T
.
Reciprocal organ interactions during heart failure: a position paper from the ESC working group on myocardial function
.
Cardiovasc Res
2021
;
117
:
2416
2433
.

136

Song
MK
,
Davies
NM
,
Roufogalis
BD
,
Huang
TH
.
Management of cardiorenal metabolic syndrome in diabetes mellitus: a phytotherapeutic perspective
.
J Diabetes Res
.
2014
;
2014
:
313718
.

137

Maaten JM
T
,
Damman
K
,
Verhaar
MC
,
Paulus
WJ
,
Duncker
DJ
,
Cheng
C
,
van Heerebeek
L
,
Hillege
HL
,
Lam
CS
,
Navis
G
,
Voors
AA
.
Connecting heart failure with preserved ejection fraction and renal dysfunction: the role of endothelial dysfunction and inflammation
.
Eur J Heart Fail
2016
;
18
:
588
598
.

138

Sarkozy
M
,
Gaspar
R
,
Zvara
A
,
Siska
A
,
Kovari
B
,
Szucs
G
,
Marvanykovi
F
,
Kovacs
MG
,
Dioszegi
P
,
Bodai
L
,
Zsindely
N
,
Pipicz
M
,
Gomori
K
,
Kiss
K
,
Bencsik
P
,
Cserni
G
,
Puskas
LG
,
Foldesi
I
,
Thum
T
,
Batkai
S
,
Csont
T
.
Chronic kidney disease induces left ventricular overexpression of the pro-hypertrophic microRNA-212
.
Sci Rep
.
2019
;
9
:
1302
.

139

Savira
F
,
Magaye
R
,
Liew
D
,
Reid
C
,
Kelly
DJ
,
Kompa
AR
,
Sangaralingham
SJ
,
Burnett
JC
, Jr.
,
Kaye
D
,
Wang
BH
.
Cardiorenal syndrome: multi-organ dysfunction involving the heart, kidney and vasculature
.
Br J Pharmacol
2020
;
177
:
2906
2922
.

140

van de Wouw
J
,
Broekhuizen
M
,
Sorop
O
,
Joles
JA
,
Verhaar
MC
,
Duncker
DJ
,
Danser
AHJ
and
Merkus
D
.
Chronic kidney disease as a risk factor for heart failure with preserved ejection fraction: a focus on microcirculatory factors and therapeutic targets
.
Front Physiol
2019
;
10
:
1108
.

141

Kumric
M
,
Ticinovic Kurir
T
,
Borovac
JA
,
Bozic
J
.
Role of novel biomarkers in diabetic cardiomyopathy
.
World J Diabetes
2021
;
12
:
685
705
.

142

Kristensen
SL
,
Rorth
R
,
Jhund
PS
,
Docherty
KF
,
Sattar
N
,
Preiss
D
,
Kober
L
,
Petrie
MC
,
McMurray
JJV
.
Cardiovascular, mortality, and kidney outcomes with GLP-1 receptor agonists in patients with type 2 diabetes: a systematic review and meta-analysis of cardiovascular outcome trials
.
Lancet Diabetes Endocrinol
2019
;
7
:
776
785
.

143

Bhatt
DL
,
Szarek
M
,
Steg
PG
,
Cannon
CP
,
Leiter
LA
,
McGuire
DK
,
Lewis
JB
,
Riddle
MC
,
Voors
AA
,
Metra
M
,
Lund
LH
,
Komajda
M
,
Testani
JM
,
Wilcox
CS
,
Ponikowski
P
,
Lopes
RD
,
Verma
S
,
Lapuerta
P
,
Pitt
B
,
Investigators
S-WT
.
Sotagliflozin in patients with diabetes and recent worsening heart failure
.
N Engl J Med
2021
;
384
:
117
128
.

144

Perkovic
V
,
Jardine
MJ
,
Neal
B
,
Bompoint
S
,
Heerspink
HJL
,
Charytan
DM
,
Edwards
R
,
Agarwal
R
,
Bakris
G
,
Bull
S
,
Cannon
CP
,
Capuano
G
,
Chu
PL
,
de Zeeuw
D
,
Greene
T
,
Levin
A
,
Pollock
C
,
Wheeler
DC
,
Yavin
Y
,
Zhang
H
,
Zinman
B
,
Meininger
G
,
Brenner
BM
,
Mahaffey
KW
,
CREDENCE Trial Investigators
.
Canagliflozin and renal outcomes in type 2 diabetes and nephropathy
.
N Engl J Med
2019
;
380
:
2295
2306
.

145

Heerspink
HJL
,
Stefansson
BV
,
Correa-Rotter
R
,
Chertow
GM
,
Greene
T
,
Hou
FF
,
Mann
JFE
,
McMurray
JJV
,
Lindberg
M
,
Rossing
P
,
Sjostrom
CD
,
Toto
RD
,
Langkilde
AM
,
Wheeler
DC
,
DAPA-CKD Trial Committees and Investigators
.
Dapagliflozin in patients with chronic kidney disease
.
N Engl J Med
2020
;
383
:
1436
1446
.

146

Braunwald
E
.
Diabetes, heart failure, and renal dysfunction: the vicious circles
.
Prog Cardiovasc Dis
2019
;
62
:
298
302
.

147

Abdesselam
I
,
Pepino
P
,
Troalen
T
,
Macia
M
,
Ancel
P
,
Masi
B
,
Fourny
N
,
Gaborit
B
,
Giannesini
B
,
Kober
F
,
Dutour
A
,
Bernard
M
.
Time course of cardiometabolic alterations in a high fat high sucrose diet mice model and improvement after GLP-1 analog treatment using multimodal cardiovascular magnetic resonance
.
J Cardiovasc Magn Reson
2015
;
17
:
95
.

148

Jonker
JT
,
de Heer
P
,
Engelse
MA
,
van Rossenberg
EH
,
Klessens
CQF
,
Baelde
HJ
,
Bajema
IM
,
Koopmans
SJ
,
Coelho
PG
,
Streefland
TCM
,
Webb
AG
,
Dekkers
IA
,
Rabelink
TJ
,
Rensen
PCN
,
Lamb
HJ
,
de Vries
APJ
.
Metabolic imaging of fatty kidney in diabesity: validation and dietary intervention
.
Nephrol Dial Transplant
2018
;
33
:
224
230
.

149

Chen
Y
,
Jiang
Z
,
Long
L
,
Miu
Y
,
Zhang
L
,
Zhong
D
,
Tang
Q
.
Magnetic resonance imaging: proton density fat fraction for assessment of pancreatic fatty infiltration during progression of T2DM bama minipigs
.
J Magn Reson Imaging
2019
;
50
:
1905
1913
.

150

Andreadou
I
,
Tsoumani
M
,
Vilahur
G
,
Ikonomidis
I
,
Badimon
L
,
Varga
ZV
,
Ferdinandy
P
,
Schulz
R
.
PCSK9 In myocardial infarction and cardioprotection: importance of lipid metabolism and inflammation
.
Front Physiol
2020
;
11
:
602497
.

151

van de Weijer
T
,
Schrauwen-Hinderling
VB
,
Schrauwen
P
.
Lipotoxicity in type 2 diabetic cardiomyopathy
.
Cardiovasc Res
2011
;
92
:
10
18
.

152

Sverdlov
AL
,
Figtree
GA
,
Horowitz
JD
,
Ngo
DT
.
Interplay between oxidative stress and inflammation in cardiometabolic syndrome
.
Mediators Inflamm
.
2016
;
2016
:
8254590
.

153

Harayama
T
,
Riezman
H
.
Understanding the diversity of membrane lipid composition
.
Nat Rev Mol Cell Biol
2018
;
19
:
281
296
.

154

Fuentes
E
,
Fuentes
F
,
Vilahur
G
,
Badimon
L
,
Palomo
I
.
Mechanisms of chronic state of inflammation as mediators that link obese adipose tissue and metabolic syndrome
.
Mediators Inflamm
2013
;
2013
:
136584
.

155

Antonopoulos
AS
,
Antoniades
C
.
The role of epicardial adipose tissue in cardiac biology: classic concepts and emerging roles
.
J Physiol
2017
;
595
:
3907
3917
.

156

Oikonomou
EK
,
Antoniades
C
.
The role of adipose tissue in cardiovascular health and disease
.
Nat Rev Cardiol
2019
;
16
:
83
99
.

157

Packer
M
.
Epicardial adipose tissue may mediate deleterious effects of obesity and inflammation on the myocardium
.
J Am Coll Cardiol
2018
;
71
:
2360
2372
.

158

Fitzgibbons
TP
,
Czech
MP
.
Epicardial and perivascular adipose tissues and their influence on cardiovascular disease: basic mechanisms and clinical associations
.
J Am Heart Assoc
2014
;
3
:
e000582
.

159

Margaritis
M
,
Antonopoulos
AS
,
Digby
J
,
Lee
R
,
Reilly
S
,
Coutinho
P
,
Shirodaria
C
,
Sayeed
R
,
Petrou
M
,
De Silva
R
,
Jalilzadeh
S
,
Demosthenous
M
,
Bakogiannis
C
,
Tousoulis
D
,
Stefanadis
C
,
Choudhury
RP
,
Casadei
B
,
Channon
KM
,
Antoniades
C
.
Interactions between vascular wall and perivascular adipose tissue reveal novel roles for adiponectin in the regulation of endothelial nitric oxide synthase function in human vessels
.
Circulation
2013
;
127
:
2209
2221
.

160

Badimon
L
,
Bugiardini
R
,
Cenko
E
,
Cubedo
J
,
Dorobantu
M
,
Duncker
DJ
,
Estruch
R
,
Milicic
D
,
Tousoulis
D
,
Vasiljevic
Z
,
Vilahur
G
,
de Wit
C
,
Koller
A
.
Position paper of the European society of cardiology-working group of coronary pathophysiology and microcirculation: obesity and heart disease
.
Eur Heart J
2017
;
38
:
1951
1958
.

161

Iozzo
P
,
Guzzardi
MA
.
Cross-talk between adipose tissue health, myocardial metabolism and vascular function: the adipose-myocardial and adipose-vascular axes
.
Curr Pharm Des
2016
;
22
:
59
67
.

162

Neeland
IJ
,
Winders
BR
,
Ayers
CR
,
Das
SR
,
Chang
AY
,
Berry
JD
,
Khera
A
,
McGuire
DK
,
Vega
GL
,
de Lemos
JA
,
Turer
AT
.
Higher natriuretic peptide levels associate with a favorable adipose tissue distribution profile
.
J Am Coll Cardiol
2013
;
62
:
752
760
.

163

Rohm
M
,
Zeigerer
A
,
Machado
J
,
Herzig
S
.
Energy metabolism in cachexia
.
EMBO Rep
2019
;
20
(4):e47258.

164

Bhatia
LS
,
Curzen
NP
,
Calder
PC
,
Byrne
CD
.
Non-alcoholic fatty liver disease: a new and important cardiovascular risk factor?
Eur Heart J
2012
;
33
:
1190
1200
.

165

Theurey
P
,
Rieusset
J
.
Mitochondria-associated membranes response to nutrient availability and role in metabolic diseases
.
Trends Endocrinol Metab
2017
;
28
:
32
45
.

166

Tubbs
E
,
Theurey
P
,
Vial
G
,
Bendridi
N
,
Bravard
A
,
Chauvin
MA
,
Ji-Cao
J
,
Zoulim
F
,
Bartosch
B
,
Ovize
M
,
Vidal
H
,
Rieusset
J
.
Mitochondria-associated endoplasmic reticulum membrane (MAM) integrity is required for insulin signaling and is implicated in hepatic insulin resistance
.
Diabetes
2014
;
63
:
3279
3294
.

167

Rashed
HM
,
Nair
BG
,
Patel
TB
.
Regulation of hepatic glycolysis and gluconeogenesis by atrial natriuretic peptide
.
Arch Biochem Biophys
1992
;
298
:
640
645
.

168

Grueter
CE
,
van Rooij
E
,
Johnson
BA
,
DeLeon
SM
,
Sutherland
LB
,
Qi
X
,
Gautron
L
,
Elmquist
JK
,
Bassel-Duby
R
,
Olson
EN
.
A cardiac microRNA governs systemic energy homeostasis by regulation of MED13
.
Cell
2012
;
149
:
671
683
.

169

Nakamura
M
,
Sadoshima
J
.
Heart over mind: metabolic control of white adipose tissue and liver
.
EMBO Mol Med
2014
;
6
:
1521
1524
.

170

Izzo
A
,
Massimino
E
,
Riccardi
G
,
Della Pepa
G
.
A narrative review on sarcopenia in type 2 diabetes mellitus: prevalencae and associated factors
.
Nutrients
2021
;
13(1):183.

171

Mesinovic
J
,
Zengin
A
,
De Courten
B
,
Ebeling
PR
,
Scott
D
.
Sarcopenia and type 2 diabetes mellitus: a bidirectional relationship
.
Diabetes Metab Syndr Obes
2019
;
12
:
1057
1072
.

172

Aubertin-Leheudre
M
,
Lord
C
,
Goulet
ED
,
Khalil
A
,
Dionne
IJ
.
Effect of sarcopenia on cardiovascular disease risk factors in obese postmenopausal women
.
Obesity
2006
;
14
:
2277
2283
.

173

Han
P
,
Yu
H
,
Ma
Y
,
Kang
L
,
Fu
L
,
Jia
L
,
Chen
X
,
Yu
X
,
Hou
L
,
Wang
L
,
Zhang
W
,
Yin
H
,
Niu
K
and
Guo
Q
.
The increased risk of sarcopenia in patients with cardiovascular risk factors in suburb-dwelling older Chinese using the AWGS definition
.
Sci Rep
2017
;
7
:
9592
.

174

Srikanthan
P
,
Hevener
AL
,
Karlamangla
AS
.
Sarcopenia exacerbates obesity-associated insulin resistance and dysglycemia: findings from the national health and nutrition examination survey III
.
PLoS One
2010
;
5
:
e10805
.

175

Garnham
JO
,
Roberts
LD
,
Espino-Gonzalez
E
,
Whitehead
A
,
Swoboda
PP
,
Koshy
A
,
Gierula
J
,
Paton
MF
,
Cubbon
RM
,
Kearney
MT
,
Egginton
S
,
Bowen
TS
,
Witte
KK
.
Chronic heart failure with diabetes mellitus is characterized by a severe skeletal muscle pathology
.
J Cachexia Sarcopenia Muscle
2020
;
11
:
394
404
.

176

Riching
AS
,
Major
JL
,
Londono
P
,
Bagchi
RA
.
The brain-heart axis: Alzheimer’s. Diabetes, and hypertension
.
ACS Pharmacol Transl Sci
2020
;
3
:
21
28
.

177

Janson
J
,
Laedtke
T
,
Parisi
JE
,
O’Brien
P
,
Petersen
RC
,
Butler
PC
.
Increased risk of type 2 diabetes in Alzheimer disease
.
Diabetes
2004
;
53
:
474
481
.

178

Campbell
JM
,
Stephenson
MD
,
de Courten
B
,
Chapman
I
,
Bellman
SM
,
Aromataris
E
.
Metformin use associated with reduced risk of dementia in patients with diabetes: a systematic review and meta-analysis
.
J Alzheimers Dis
2018
;
65
:
1225
1236
.

179

Nguyen
BY
,
Azam
T
,
Wang
X
.
Cellular signaling cross-talk between different cardiac cell populations: an insight into the role of exosomes in the heart diseases and therapy
.
Am J Physiol Heart Circ Physiol
2021
;
320
:
H1213
H1234
.

180

Russo
I
,
Frangogiannis
NG
.
Diabetes-associated cardiac fibrosis: cellular effectors, molecular mechanisms and therapeutic opportunities
.
J Mol Cell Cardiol
2016
;
90
:
84
93
.

181

Neumann
S
,
Huse
K
,
Semrau
R
,
Diegeler
A
,
Gebhardt
R
,
Buniatian
GH
,
Scholz
GH
.
Aldosterone and D-glucose stimulate the proliferation of human cardiac myofibroblasts in vitro
.
Hypertension
2002
;
39
:
756
760
.

182

Tokudome
T
,
Horio
T
,
Yoshihara
F
,
Suga
S
,
Kawano
Y
,
Kohno
M
,
Kangawa
K
.
Direct effects of high glucose and insulin on protein synthesis in cultured cardiac myocytes and DNA and collagen synthesis in cardiac fibroblasts
.
Metab Clin Exp
2004
;
53
:
710
715
.

183

Asbun
J
,
Manso
AM
,
Villarreal
FJ
.
Profibrotic influence of high glucose concentration on cardiac fibroblast functions: effects of losartan and vitamin E
.
Am J Physiol Heart Circ Physiol
2005
;
288
:
H227
H234
.

184

Civitarese
RA
,
Talior-Volodarsky
I
,
Desjardins
JF
,
Kabir
G
,
Switzer
J
,
Mitchell
M
,
Kapus
A
,
McCulloch
CA
,
Gullberg
D
,
Connelly
KA
.
The alpha11 integrin mediates fibroblast-extracellular matrix-cardiomyocyte interactions in health and disease
.
Am J Physiol Heart Circ Physiol
2016
;
311
:
H96
H106
.

185

Sandesara
PB
,
O'Neal
WT
,
Kelli
HM
,
Samman-Tahhan
A
,
Hammadah
M
,
Quyyumi
AA
,
Sperling
LS
.
The prognostic significance of diabetes and microvascular complications in patients with heart failure with preserved ejection fraction
.
Diabetes Care
2018
;
41
:
150
155
.

186

Lam
CS
,
Lund
LH
.
Microvascular endothelial dysfunction in heart failure with preserved ejection fraction
.
Heart
2016
;
102
:
257
259
.

187

Bendall
JK
,
Douglas
G
,
McNeill
E
,
Channon
KM
,
Crabtree
MJ
.
Tetrahydrobiopterin in cardiovascular health and disease
.
Antioxid Redox Signal
2014
;
20
:
3040
3077
.

188

Hopf
AE
,
Andresen
C
,
Kotter
S
,
Isic
M
,
Ulrich
K
,
Sahin
S
,
Bongardt
S
,
Roll
W
,
Drove
F
,
Scheerer
N
,
Vandekerckhove
L
,
De Keulenaer
GW
,
Hamdani
N
,
Linke
WA
,
Kruger
M
.
Diabetes-induced cardiomyocyte passive stiffening is caused by impaired insulin-dependent titin modification and can be modulated by neuregulin-1
.
Circ Res
2018
;
123
:
342
355
.

189

Travers
JG
,
Kamal
FA
,
Robbins
J
,
Yutzey
KE
,
Blaxall
BC
.
Cardiac fibrosis: the fibroblast awakens
.
Circ Res
2016
;
118
:
1021
1040
.

190

Zhou
T
,
Hu
Z
,
Yang
S
,
Sun
L
,
Yu
Z
,
Wang
G
.
Role of adaptive and innate immunity in type 2 diabetes mellitus
.
J Diabetes Res
2018
;
2018
:
7457269
.

191

Bajpai
A
,
Tilley
DG
.
The role of leukocytes in diabetic cardiomyopathy
.
Front Physiol
2018
;
9
:
1547
.

192

Abdullah
CS
,
Li
Z
,
Wang
X
,
Jin
ZQ
.
Depletion of T lymphocytes ameliorates cardiac fibrosis in streptozotocin-induced diabetic cardiomyopathy
.
Int Immunopharmacol
2016
;
39
:
251
264
.

193

Laroumanie
F
,
Douin-Echinard
V
,
Pozzo
J
,
Lairez
O
,
Tortosa
F
,
Vinel
C
,
Delage
C
,
Calise
D
,
Dutaur
M
,
Parini
A
,
Pizzinat
N
.
CD4+ T cells promote the transition from hypertrophy to heart failure during chronic pressure overload
.
Circulation
2014
;
129
:
2111
2114
.

194

Zhao
RX
,
Li
WJ
,
Lu
YR
,
Qin
J
,
Wu
CL
,
Tian
M
,
He
TY
,
Yi
SN
,
Tang
DQ
,
Sun
L
,
Chen
L
.
Increased peripheral proinflammatory T helper subsets contribute to cardiovascular complications in diabetic patients
.
Mediators Inflamm
2014
;
2014
:
596967
.

195

Tang
H
,
Zhong
Y
,
Zhu
Y
,
Zhao
F
,
Cui
X
,
Wang
Z
.
Low responder T cell susceptibility to the suppressive function of regulatory T cells in patients with dilated cardiomyopathy
.
Heart
2010
;
96
:
765
771
.

196

Huang
X
,
Qin
Z
,
Xu
M
,
Zhang
F
,
Jiang
X
,
Hua
F
,
Tao
L
.
Neutrophil: lymphocyte ratio is positively associated with subclinical diabetic cardiomyopathy
.
BMC Endocr Disord
2020
;
20
:
99
.

197

Garris
CS
,
Blaho
VA
,
Hla
T
,
Han
MH
.
Sphingosine-1-phosphate receptor 1 signalling in T cells: trafficking and beyond
.
Immunology
2014
;
142
:
347
353
.

198

Abdullah
CS
,
Jin
ZQ
.
Targeted deletion of T-cell S1P receptor 1 ameliorates cardiac fibrosis in streptozotocin-induced diabetic mice
.
FASEB J
2018
;
32
:
5426
5435
.

199

DeBerge
M
,
Zhang
S
,
Glinton
K
,
Grigoryeva
L
,
Hussein
I
,
Vorovich
E
,
Ho
K
,
Luo
X
,
Thorp
EB
.
Efferocytosis and outside-in signaling by cardiac phagocytes. Links to repair, cellular programming, and intercellular crosstalk in heart
.
Front Immunol
2017
;
8
:
1428
.

200

Babu S
S
,
Thandavarayan
RA
,
Joladarashi
D
,
Jeyabal
P
,
Krishnamurthy
S
,
Bhimaraj
A
,
Youker
KA
,
Krishnamurthy
P
.
MicroRNA-126 overexpression rescues diabetes-induced impairment in efferocytosis of apoptotic cardiomyocytes
.
Sci Rep
2016
;
6
:
36207
.

201

Hulsmans
M
,
Sager
HB
,
Roh
JD
,
Valero-Munoz
M
,
Houstis
NE
,
Iwamoto
Y
,
Sun
Y
,
Wilson
RM
,
Wojtkiewicz
G
,
Tricot
B
,
Osborne
MT
,
Hung
J
,
Vinegoni
C
,
Naxerova
K
,
Sosnovik
DE
,
Zile
MR
,
Bradshaw
AD
,
Liao
R
,
Tawakol
A
,
Weissleder
R
,
Rosenzweig
A
,
Swirski
FK
,
Sam
F
,
Nahrendorf
M
.
Cardiac macrophages promote diastolic dysfunction
.
J Exp Med
2018
;
215
:
423
440
.

202

Jia
G
,
Hill
MA
,
Sowers
JR
.
Diabetic cardiomyopathy: an update of mechanisms contributing to this clinical entity
.
Circ Res
2018
;
122
:
624
638
.

203

Sowton
AP
,
Griffin
JL
,
Murray
AJ
.
Metabolic profiling of the diabetic heart: toward a richer picture
.
Front Physiol
2019
;
10
:
639
.

204

Abdurrachim
D
,
Nabben
M
,
Hoerr
V
,
Kuhlmann
MT
,
Bovenkamp
P
,
Ciapaite
J
,
Geraets
IME
,
Coumans
W
,
Luiken
J
,
Glatz
JFC
,
Schafers
M
,
Nicolay
K
,
Faber
C
,
Hermann
S
,
Prompers
JJ
.
Diabetic db/db mice do not develop heart failure upon pressure overload: a longitudinal in vivo PET, MRI, and MRS study on cardiac metabolic, structural, and functional adaptations
.
Cardiovasc Res
2017
;
113
:
1148
1160
.

205

Bollano
E
,
Omerovic
E
,
Svensson
H
,
Waagstein
F
,
Fu
M
.
Cardiac remodeling rather than disturbed myocardial energy metabolism is associated with cardiac dysfunction in diabetic rats
.
Int J Cardiol
2007
;
114
:
195
201
.

206

Abdurrachim
D
,
Ciapaite
J
,
Wessels
B
,
Nabben
M
,
Luiken
JJ
,
Nicolay
K
,
Prompers
JJ
.
Cardiac diastolic dysfunction in high-fat diet fed mice is associated with lipotoxicity without impairment of cardiac energetics in vivo
.
Biochim Biophys Acta
2014
;
1842
:
1525
1537
.

207

Johnson
R
,
Nxele
X
,
Cour
M
,
Sangweni
N
,
Jooste
T
,
Hadebe
N
,
Samodien
E
,
Benjeddou
M
,
Mazino
M
,
Louw
J
,
Lecour
S
.
Identification of potential biomarkers for predicting the early onset of diabetic cardiomyopathy in a mouse model
.
Sci Rep
2020
;
10
:
12352
.

208

Renner
S
,
Blutke
A
,
Clauss
S
,
Deeg
CA
,
Kemter
E
,
Merkus
D
,
Wanke
R
,
Wolf
E
.
Porcine models for studying complications and organ crosstalk in diabetes mellitus
.
Cell Tissue Res
2020
;
380
:
341
378
.

209

Jiang
B
,
Guo
N
,
Ge
Y
,
Zhang
L
,
Oudkerk
M
,
Xie
X
.
Development and application of artificial intelligence in cardiac imaging
.
Br J Radiol
2020
;
93
:
20190812
.

210

Chatham
JC
,
Young
ME
,
Zhang
J
.
Role of O-linked N-acetylglucosamine (O-GlcNAc) modification of proteins in diabetic cardiovascular complications
.
Curr Opin Pharmacol
2021
;
57
:
1
12
.

211

Bodiga
VL
,
Eda
SR
,
Bodiga
S
.
Advanced glycation end products: role in pathology of diabetic cardiomyopathy
.
Heart Fail Rev
2014
;
19
:
49
63
.

212

Byrne
NJ
,
Rajasekaran
NS
,
Abel
ED
,
Bugger
H
.
Therapeutic potential of targeting oxidative stress in diabetic cardiomyopathy
.
Free Radic Biol Med
2021
;
169
:
317
342
.

213

McDonagh
TA
,
Metra
M
,
Adamo
M
,
Gardner
RS
,
Baumbach
A
,
Bohm
M
,
Burri
H
,
Butler
J
,
Celutkiene
J
,
Chioncel
O
,
Cleland
JGF
,
Coats
AJS
,
Crespo-Leiro
MG
,
Farmakis
D
,
Gilard
M
,
Heymans
S
,
Hoes
AW
,
Jaarsma
T
,
Jankowska
EA
,
Lainscak
M
,
Lam
CSP
,
Lyon
AR
,
McMurray
JJV
,
Mebazaa
A
,
Mindham
R
,
Muneretto
C
,
Francesco Piepoli
M
,
Price
S
,
Rosano
GMC
,
Ruschitzka
F
,
Kathrine Skibelund
A
,
ESC Scientific Document Group
.
2021 ESC guidelines for the diagnosis and treatment of acute and chronic heart failure
.
Eur Heart J
2021
;
42
:
3599
3726
.

214

Joshi
SS
,
Singh
T
,
Newby
DE
,
Singh
J
.
Sodium-glucose co-transporter 2 inhibitor therapy: mechanisms of action in heart failure
.
Heart
2021
; 107(13):1032–8.

215

Zelniker
TA
,
Braunwald
E
.
Mechanisms of cardiorenal effects of sodium-glucose cotransporter 2 inhibitors: JACC state-of-the-art review
.
J Am Coll Cardiol
2020
;
75
:
422
434
.

216

Seferovic
PM
,
Fragasso
G
,
Petrie
M
,
Mullens
W
,
Ferrari
R
,
Thum
T
,
Bauersachs
J
,
Anker
SD
,
Ray
R
,
Cavusoglu
Y
,
Polovina
M
,
Metra
M
,
Ambrosio
G
,
Prasad
K
,
Seferovic
J
,
Jhund
PS
,
Dattilo
G
,
Celutkiene
J
,
Piepoli
M
,
Moura
B
,
Chioncel
O
,
Ben Gal
T
,
Heymans
S
,
de Boer
RA
,
Jaarsma
T
,
Hill
L
,
Lopatin
Y
,
Lyon
AR
,
Ponikowski
P
,
Lainscak
M
,
Jankowska
E
,
Mueller
C
,
Cosentino
F
,
Lund
L
,
Filippatos
GS
,
Ruschitzka
F
,
Coats
AJS
,
Rosano
GMC
.
Sodium-glucose co-transporter 2 inhibitors in heart failure: beyond glycaemic control. A position paper of the Heart Failure Association of the European Society of Cardiology
.
Eur J Heart Fail
2020
;
22
:
1495
1503
.

217

Lee
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
.

218

Yurista
SR
,
Sillje
HHW
,
Oberdorf-Maass
SU
,
Schouten
EM
,
Pavez Giani
MG
,
Hillebrands
JL
,
van Goor
H
,
van Veldhuisen
DJ
,
de Boer
RA
,
Westenbrink
BD
.
Sodium-glucose co-transporter 2 inhibition with empagliflozin improves cardiac function in non-diabetic rats with left ventricular dysfunction after myocardial infarction
.
Eur J Heart Fail
2019
;
21
:
862
873
.

219

Uthman
L
,
Baartscheer
A
,
Bleijlevens
B
,
Schumacher
CA
,
Fiolet
JWT
,
Koeman
A
,
Jancev
M
,
Hollmann
MW
,
Weber
NC
,
Coronel
R
,
Zuurbier
CJ
.
Class effects of SGLT2 inhibitors in mouse cardiomyocytes and hearts: inhibition of na(+)/H(+) exchanger, lowering of cytosolic na(+) and vasodilation
.
Diabetologia
2018
;
61
:
722
726
.

220

Bugger
H
,
Abel
ED
.
Rodent models of diabetic cardiomyopathy
.
Dis Model Mech
2009
;
2
:
454
466
.

221

Ouwens
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
.

222

Tschope
C
,
Walther
T
,
Escher
F
,
Spillmann
F
,
Du
J
,
Altmann
C
,
Schimke
I
,
Bader
M
,
Sanchez-Ferrer
CF
,
Schultheiss
HP
,
Noutsias
M
.
Transgenic activation of the kallikrein-kinin system inhibits intramyocardial inflammation, endothelial dysfunction and oxidative stress in experimental diabetic cardiomyopathy
.
FASEB J
2005
;
19
:
2057
2059
.

223

Fillmore
N
,
Wagg
CS
,
Zhang
L
,
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
.

224

Shao
D
,
Kolwicz
SC
Jr
,
Wang
P
,
Roe
ND
,
Villet
O
,
Nishi
K
,
Hsu
YA
,
Flint
GV
,
Caudal
A
,
Wang
W
,
Regnier
M
,
Tian
R
.
Increasing fatty acid oxidation prevents high-fat diet-induced cardiomyopathy through regulating parkin-mediated mitophagy
.
Circulation
.
2020
;
142
:
983
997
.

225

Zhong
P
,
Quan
D
,
Huang
Y
,
Huang
H
.
CaMKII activation promotes cardiac electrical remodeling and increases the susceptibility to arrhythmia induction in high-fat diet-fed mice with hyperlipidemia conditions
.
J Cardiovasc Pharmacol
2017
;
70
:
245
254
.

226

Wang
Q
,
Wang
Y
,
West
TM
,
Liu
Y
,
Reddy
GR
,
Barbagallo
F
,
Xu
B
,
Shi
Q
,
Deng
B
,
Wei
W
,
Xiang
YK
.
Carvedilol induces biased beta1 adrenergic receptor-nitric oxide synthase 3-cyclic guanylyl monophosphate signalling to promote cardiac contractility
.
Cardiovasc Res
2021
;
117
:
2237
2251
.

227

Zhang
Y
,
Bao
M
,
Dai
M
,
Wang
X
,
He
W
,
Tan
T
,
Lin
D
,
Wang
W
,
Wen
Y
,
Zhang
R
.
Cardiospecific CD36 suppression by lentivirus-mediated RNA interference prevents cardiac hypertrophy and systolic dysfunction in high-fat-diet induced obese mice
.
Cardiovasc Diabetol
2015
;
14
:
69
.

228

Li
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
.

229

Mazumder
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
.

230

Boudina
S
,
Sena
S
,
O’Neill
BT
,
Tathireddy
P
,
Young
ME
,
Abel
ED
.
Reduced mitochondrial oxidative capacity and increased mitochondrial uncoupling impair myocardial energetics in obesity
.
Circulation
2005
;
112
:
2686
2695
.

231

An
HS
,
Lee
JY
,
Choi
EB
,
Jeong
EA
,
Shin
HJ
,
Kim
KE
,
Park
KA
,
Jin
Z
,
Lee
JE
,
Koh
JS
,
Kwak
W
,
Kim
WH
,
Roh
GS
.
Caloric restriction reverses left ventricular hypertrophy through the regulation of cardiac iron homeostasis in impaired leptin signaling mice
.
Sci Rep
2020
;
10
:
7176
.

232

Barouch
LA
,
Gao
D
,
Chen
L
,
Miller
KL
,
Xu
W
,
Phan
AC
,
Kittleson
MM
,
Minhas
KM
,
Berkowitz
DE
,
Wei
C
,
Hare
JM
.
Cardiac myocyte apoptosis is associated with increased DNA damage and decreased survival in murine models of obesity
.
Circ Res
2006
;
98
:
119
124
.

233

Finck
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 U S A
2003
;
100
:
1226
1231
.

234

Flarsheim
CE
,
Grupp
IL
,
Matlib
MA
.
Mitochondrial dysfunction accompanies diastolic dysfunction in diabetic rat heart
.
Am J Physiol
1996
;
271
:
H192
H202
.

235

Popovich
BK
,
Boheler
KR
,
Dillmann
WH
.
Diabetes decreases creatine kinase enzyme activity and mRNA level in the rat heart
.
Am J Physiol
1989
;
257
:
E573
E577
.

236

Wold
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
.

237

Santos
DL
,
Palmeira
CM
,
Seica
R
,
Dias
J
,
Mesquita
J
,
Moreno
AJ
,
Santos
MS
.
Diabetes and mitochondrial oxidative stress: a study using heart mitochondria from the diabetic goto-kakizaki rat
.
Mol Cell Biochem
2003
;
246
:
163
170
.

238

Tikellis
C
,
Thomas
MC
,
Harcourt
BE
,
Coughlan
MT
,
Pete
J
,
Bialkowski
K
,
Tan
A
,
Bierhaus
A
,
Cooper
ME
,
Forbes
JM
.
Cardiac inflammation associated with a western diet is mediated via activation of RAGE by AGEs
.
Am J Physiol Endocrinol Metab
2008
;
295
:
E323
E330
.

239

Liu
X
,
Liu
S
.
Role of microRNAs in the pathogenesis of diabetic cardiomyopathy
.
Biomed Rep
2017
;
6
:
140
145
.

240

Zheng
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
.

241

Umbarawan
Y
,
Kawakami
R
,
Syamsunarno
M
,
Koitabashi
N
,
Obinata
H
,
Yamaguchi
A
,
Hanaoka
H
,
Hishiki
T
,
Hayakawa
N
,
Sunaga
H
,
Matsui
H
,
Kurabayashi
M
,
Iso
T
.
Reduced fatty acid uptake aggravates cardiac contractile dysfunction in streptozotocin-induced diabetic cardiomyopathy
.
Sci Rep
2020
;
10
:
20809
.

242

Roslan
J
,
Giribabu
N
,
Karim
K
,
Salleh
N
.
Quercetin ameliorates oxidative stress, inflammation and apoptosis in the heart of streptozotocin-nicotinamide-induced adult male diabetic rats
.
Biomed Pharmacother
2017
;
86
:
570
582
.

243

Soliman
H
,
Nyamandi
V
,
Garcia-Patino
M
,
Zhang
PC
,
Lin
E
,
Jia
ZP
,
Tibbits
GF
,
Hove-Madsen
L
,
MacLeod
KM
.
ROCK2 Promotes ryanodine receptor phosphorylation and arrhythmic calcium release in diabetic cardiomyocytes
.
Int J Cardiol
2019
;
281
:
90
98
.

244

Boudina
S
,
Abel
ED
.
Diabetic cardiomyopathy revisited
.
Circulation
2007
;
115
:
3213
3223
.

245

Buchanan
J
,
Mazumder
PK
,
Hu
P
,
Chakrabarti
G
,
Roberts
MW
,
Yun
UJ
,
Cooksey
RC
,
Litwin
SE
and
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
.

246

Mori
J
,
Patel
VB
,
Abo Alrob
O
,
Basu
R
,
Altamimi
T
,
Desaulniers
J
,
Wagg
CS
,
Kassiri
Z
,
Lopaschuk
GD
,
Oudit
GY
.
Angiotensin 1-7 ameliorates diabetic cardiomyopathy and diastolic dysfunction in db/db mice by reducing lipotoxicity and inflammation
.
Circ Heart Fail
2014
;
7
:
327
339
.

247

Belke
DD
,
Swanson
EA
,
Dillmann
WH
.
Decreased sarcoplasmic reticulum activity and contractility in diabetic db/db mouse heart
.
Diabetes
2004
;
53
:
3201
3208
.

248

Wang
T
,
Li
J
,
Li
H
,
Zhong
X
,
Wang
L
,
Zhao
S
,
Liu
X
,
Huang
Z
,
Wang
Y
.
Aerobic exercise inhibited p2x7 purinergic receptors to improve cardiac remodeling in mice with type 2 diabetes
.
Front Physiol
2022
;
13
:
828020
.

249

Zhao
XY
,
Hu
SJ
,
Li
J
,
Mou
Y
,
Chen
BP
,
Xia
Q
.
Decreased cardiac sarcoplasmic reticulum Ca2+ -ATPase activity contributes to cardiac dysfunction in streptozotocin-induced diabetic rats
.
J Physiol Biochem
2006
;
62
:
1
8
.

250

Aragno
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
.

251

Luo
B
,
Li
B
,
Wang
W
,
Liu
X
,
Xia
Y
,
Zhang
C
,
Zhang
M
,
Zhang
Y
,
An
F
.
NLRP3 Gene silencing ameliorates diabetic cardiomyopathy in a type 2 diabetes rat model
.
PLoS One
2014
;
9
:
e104771
.

252

Christe
ME
,
Rodgers
RL
.
Cardiac glucose and fatty acid oxidation in the streptozotocin-induced diabetic spontaneously hypertensive rat
.
Hypertension
1995
;
25
:
235
241
.

253

Ferreira
R
,
Guerra
G
,
Padrao
AI
,
Melo
T
,
Vitorino
R
,
Duarte
JA
,
Remiao
F
,
Domingues
P
,
Amado
F
,
Domingues
MR
.
Lipidomic characterization of streptozotocin-induced heart mitochondrial dysfunction
.
Mitochondrion
2013
;
13
:
762
771
.

254

Yoon
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
.

255

Chatham
JC
,
Seymour
AM
.
Cardiac carbohydrate metabolism in Zucker diabetic fatty rats
.
Cardiovasc Res
2002
;
55
:
104
112
.

256

Wang
P
,
Lloyd
SG
,
Zeng
H
,
Bonen
A
,
Chatham
JC
.
Impact of altered substrate utilization on cardiac function in isolated hearts from Zucker diabetic fatty rats
.
Am J Physiol Heart Circ Physiol
2005
;
288
:
H2102
H2110
.

257

Golfman
LS
,
Wilson
CR
,
Sharma
S
,
Burgmaier
M
,
Young
ME
,
Guthrie
PH
,
Van Arsdall
M
,
Adrogue
JV
,
Brown
KK
,
Taegtmeyer
H
.
Activation of PPARgamma enhances myocardial glucose oxidation and improves contractile function in isolated working hearts of ZDF rats
.
Am J Physiol Endocrinol Metab
2005
;
289
:
E328
E336
.

258

Jadhav
A
,
Tiwari
S
,
Lee
P
,
Ndisang
JF
.
The heme oxygenase system selectively enhances the anti-inflammatory macrophage-M2 phenotype, reduces pericardial adiposity, and ameliorated cardiac injury in diabetic cardiomyopathy in Zucker diabetic fatty rats
.
J Pharmacol Exp Ther
2013
;
345
:
239
249
.

259

Matyas
C
,
Nemeth
BT
,
Olah
A
,
Torok
M
,
Ruppert
M
,
Kellermayer
D
,
Barta
BA
,
Szabo
G
,
Kokeny
G
,
Horvath
EM
,
Bodi
B
,
Papp
Z
,
Merkely
B
,
Radovits
T
.
Prevention of the development of heart failure with preserved ejection fraction by the phosphodiesterase-5A inhibitor vardenafil in rats with type 2 diabetes
.
Eur J Heart Fail
2017
;
19
:
326
336
.

260

Beaudoin
MS
,
Perry
CG
,
Arkell
AM
,
Chabowski
A
,
Simpson
JA
,
Wright
DC
,
Holloway
GP
.
Impairments in mitochondrial palmitoyl-CoA respiratory kinetics that precede development of diabetic cardiomyopathy are prevented by resveratrol in ZDF rats
.
J Physiol
2014
;
592
:
2519
2533
.

261

Raza
H
,
John
A
,
Howarth
FC
.
Alterations in glutathione redox metabolism, oxidative stress, and mitochondrial function in the left ventricle of elderly Zucker diabetic fatty rat heart
.
Int J Mol Sci
2012
;
13
:
16241
16254
.

262

Darmellah
A
,
Baetz
D
,
Prunier
F
,
Tamareille
S
,
Rucker-Martin
C
,
Feuvray
D
.
Enhanced activity of the myocardial na+/H+ exchanger contributes to left ventricular hypertrophy in the goto-kakizaki rat model of type 2 diabetes: critical role of AKT
.
Diabetologia
2007
;
50
:
1335
1344
.

263

Bugger
H
,
Riehle
C
,
Jaishy
B
,
Wende
AR
,
Tuinei
J
,
Chen
D
,
Soto
J
,
Pires
KM
,
Boudina
S
,
Theobald
HA
,
Luptak
I
,
Wayment
B
,
Wang
X
,
Litwin
SE
,
Weimer
BC
,
Abel
ED
.
Genetic loss of insulin receptors worsens cardiac efficiency in diabetes
.
J Mol Cell Cardiol
2012
;
52
:
1019
1026
.

264

Picatoste
B
,
Ramirez
E
,
Caro-Vadillo
A
,
Iborra
C
,
Ares-Carrasco
S
,
Egido
J
,
Tunon
J
,
Lorenzo
O
.
Sitagliptin reduces cardiac apoptosis, hypertrophy and fibrosis primarily by insulin-dependent mechanisms in experimental type-II diabetes. Potential roles of GLP;1, isoforms
.
PLoS One
2013
;
8
:
e78330
.

265

Devanathan
S
,
Nemanich
ST
,
Kovacs
A
,
Fettig
N
,
Gropler
RJ
,
Shoghi
KI
.
Genomic and metabolic disposition of non-obese type 2 diabetic rats to increased myocardial fatty acid metabolism
.
PLoS One
2013
;
8
:
e78477
.

266

Salem
KA
,
Adrian
TE
,
Qureshi
MA
,
Parekh
K
,
Oz
M
,
Howarth
FC
.
Shortening and intracellular Ca2 + in ventricular myocytes and expression of genes encoding cardiac muscle proteins in early onset type 2 diabetic goto-kakizaki rats
.
Exp Physiol
2012
;
97
:
1281
1291
.

267

Fuentes-Antras
J
,
Picatoste
B
,
Gomez-Hernandez
A
,
Egido
J
,
Tunon
J
,
Lorenzo
O
.
Updating experimental models of diabetic cardiomyopathy
.
J Diabetes Res
2015
;
2015
:
656795
.

268

Seal
SV
,
Henry
M
,
Pajot
C
,
Holuka
C
,
Bailbe
D
,
Movassat
J
,
Darnaudery
M
,
Turner
JD
.
A holistic view of the goto-kakizaki rat immune system: decreased circulating immune markers in non- obese type 2 diabetes
.
Front Immunol
2022
;
13
:
896179
.

269

During
MJ
,
Cao
L
,
Zuzga
DS
,
Francis
JS
,
Fitzsimons
HL
,
Jiao
X
,
Bland
RJ
,
Klugmann
M
,
Banks
WA
,
Drucker
DJ
,
Haile
CN
.
Glucagon-like peptide-1 receptor is involved in learning and neuroprotection
.
Nat Med
2003
;
9
:
1173
1179
.

Author notes

Frank Lezoualc’h and Lina Badimon contributed equally to the work.

Conflict of interest L.B. declares to have acted as SAB member of Sanofi, Novartis and International Aspirin Foundation and a Research Grant from AstraZeneca to the institution (unrelated to this work). LB and GV have founded two spinoff companies, Glycardial Diagnostics and Ivestatin Therapeutics (unrelated to this work). RAB reports speaker fees from Abbott, AstraZeneca, Bayer, Novartis, and Roche, outside the submitted work. The UMCG, which employs RAB has received research grants and/or fees from AstraZeneca, Abbott, Boehringer-Ingelheim, Cardior Pharmaceuticals Gmbh, Ionis Pharmaceuticals, Inc., Novo Nordisk, and Roche, outside the submitted work. GC, TD’H, MK and GAD are supported by the RHU-CARMMA Grant (ANR-15-RHUS-0003), the ANR-18-EURE-0011 Grant (EUR LIVE), the ANR-21-CE14-0031-01 Grant (DOXEPISEN), the RSE20003DDA Grant (FHU-SENEC), the FRM grants (EQU202003010186 and ENV202004011730). No others declared any conflict of interest.

This article is published and distributed under the terms of the Oxford University Press, Standard Journals Publication Model (https://academic.oup.com/pages/standard-publication-reuse-rights)