You are on page 1of 15

Review

Diabetic Cardiomyopathy
An Update of Mechanisms Contributing to This Clinical Entity
Guanghong Jia, Michael A. Hill, James R. Sowers

Abstract: Heart failure and related morbidity and mortality are increasing at an alarming rate, in large part,
because of increases in aging, obesity, and diabetes mellitus. The clinical outcomes associated with heart failure
are considerably worse for patients with diabetes mellitus than for those without diabetes mellitus. In people with
diabetes mellitus, the presence of myocardial dysfunction in the absence of overt clinical coronary artery disease,
valvular disease, and other conventional cardiovascular risk factors, such as hypertension and dyslipidemia,
has led to the descriptive terminology, diabetic cardiomyopathy. The prevalence of diabetic cardiomyopathy is
increasing in parallel with the increase in diabetes mellitus. Diabetic cardiomyopathy is initially characterized by
myocardial fibrosis, dysfunctional remodeling, and associated diastolic dysfunction, later by systolic dysfunction,
and eventually by clinical heart failure. Impaired cardiac insulin metabolic signaling, mitochondrial dysfunction,
increases in oxidative stress, reduced nitric oxide bioavailability, elevations in advanced glycation end products
and collagen-based cardiomyocyte and extracellular matrix stiffness, impaired mitochondrial and cardiomyocyte
calcium handling, inflammation, renin–angiotensin–aldosterone system activation, cardiac autonomic neuropathy,
endoplasmic reticulum stress, microvascular dysfunction, and a myriad of cardiac metabolic abnormalities have
all been implicated in the development and progression of diabetic cardiomyopathy. Molecular mechanisms
linked to the underlying pathophysiological changes include abnormalities in AMP-activated protein kinase,
peroxisome proliferator-activated receptors, O-linked N-acetylglucosamine, protein kinase C, microRNA, and
exosome pathways. The aim of this review is to provide a contemporary view of these instigators of diabetic
cardiomyopathy, as well as mechanistically based strategies for the prevention and treatment of diabetic
cardiomyopathy.   (Circ Res. 2018;122:624-638. DOI: 10.1161/CIRCRESAHA.117.311586.)
Key Words: diabetes mellitus ◼ heart failure ◼ myocytes, cardiac ◼ peroxisome proliferator-activated receptors
Downloaded from http://ahajournals.org by on February 10, 2022

◼ renin-angiotensin system

D iabetic cardiomyopathy is defined by the existence of abnor-


mal myocardial structure and performance in the absence
of other cardiac risk factors, such as coronary artery disease, hy-
by structural and functional abnormalities, including left ven-
tricular (LV) hypertrophy, fibrosis, and cell signaling abnormali-
ties. These pathophysiological changes of cardiac fibrosis and
pertension, and significant valvular disease, in individuals with stiffness and associated subclinical diastolic dysfunction often
diabetes mellitus. It was first described in 19721 in postmortem evolve to heart failure with normal ejection fraction and eventual
pathological findings from 4 diabetic patients who manifested systolic dysfunction accompanied by heart failure with reduced
heart failure symptoms without evidence of coronary artery ejection fraction. This review summarizes recent research explor-
or valve disease and further confirmed in a 1974 Framingham ing molecular mechanisms, structural and functional changes,
Heart Study that demonstrated a higher incidence of heart fail- and possible therapeutic approaches for the prevention and treat-
ure in diabetic women (5-fold) and men (2.4-fold) after adjust-
ment of diabetic cardiomyopathy. It also highlights unmet needs
ment for other risk factors, such as age, coronary heart disease,
and future research directions to better understand fundamental
and hypertension.2 In 2013, the American College of Cardiology
molecular abnormalities that promote this cardiomyopathy.
Foundation, the American Heart Association,3 and the European
Society of Cardiology in collaboration with the European
Association for the Study of Diabetes4 defined diabetic cardiomy- Clinical Aspects of Diabetic Cardiomyopathy
opathy as a clinical condition of ventricular dysfunction that oc- Epidemiology of Diabetes Mellitus–Related Heart
curs in the absence of coronary atherosclerosis and hypertension Failure
in patients with diabetes mellitus. In its early stages, diabetic car- Clinical trials show the prevalence of heart failure in diabetic
diomyopathy includes a hidden subclinical period characterized patients to range from 19% to 26%.5–7 The Framingham Heart

From the Diabetes and Cardiovascular Research Center (G.J., J.R.S.) and Department of Medical Pharmacology and Physiology (M.A.H., J.R.S.),
University of Missouri School of Medicine, Columbia; Dalton Cardiovascular Research Center, University of Missouri, Columbia (M.A.H., J.R.S.); and
Research Service, Truman Memorial Veterans Hospital, Columbia, MO (G.J., J.R.S.).
Correspondence to James R. Sowers, MD, or Guanghong Jia, PhD, Diabetes and Cardiovascular Research Center, University of Missouri School of
Medicine, D109 Diabetes Center HSC, One Hospital Dr, Columbia, MO 65212, E-mail Sowersj@health.missouri.edu or Jiag@health.missouri.edu
© 2018 American Heart Association, Inc.
Circulation Research is available at http://circres.ahajournals.org DOI: 10.1161/CIRCRESAHA.117.311586

624
Jia et al   Diabetic Cardiomyopathy and Heart Failure   625

Nonstandard Abbreviations and Acronyms


Risk Factors for Diabetic Cardiomyopathy
Hyperglycemia, systemic insulin resistance, and impaired
AGE advanced glycation end products cardiac insulin metabolic signaling are major clinical ab-
Akt protein kinase B normalities in diabetes mellitus, and all are involved in the
AMPK AMP-activated protein kinase pathogenesis of diabetic cardiomyopathy (Figures 1 and 2).13
Ca2+ calcium In a prospective national survey of patients with heart failure,
CD36 cluster of differentiation 36
1811 individuals with and 2182 without pre-existing diabetes
CoA coenzyme A
mellitus, glucose levels of 110 to 140, 140 to 200, and ≥200
mg/dL were associated with a 9%, 16%, and 53% increased
CREMc yclic adenosine 5′-monophosphate-responsive element
modulator mortality risk when compared with an admission blood glu-
CVD cardiovascular disease cose <110 mg/dL in patients with no pre-existing diabetes
eNOS endothelial nitric oxide synthase mellitus. There was a linear relationship between blood glu-
Erk1/2 extracellular signal-regulated kinase 1/2
cose level and long-term mortality in heart failure even in
FFA free fatty acid
patients without a clinical diagnosis of diabetes mellitus.
However, increased mortality risk was seen only in diabetics
GLUT4 glucose transporter type 4
with glucose levels >200 mg/dL.14 In the UKPDS (The UK
IRS insulin receptor substrate
Prospective Diabetes Study) clinical study, a 1% reduction
JNK c-Jun N terminal kinase
in hemoglobin A1c was associated with a 16% risk reduction
LV left ventricular
for development of heart failure,12 suggesting that there is a
MAPK mitogen-activated protein kinase
time-related log-linear relationship between long-term glyce-
miRNA microRNAs mic control and heart failure risk. Furthermore, after 4 years
MR mineralocorticoid receptors of follow-up, a community-based study of 6814 people with
mTOR mammalian target of rapamycin no initial coronary artery disease demonstrated that increas-
NF-κB nuclear factor κ-light-chain-enhancer of activated B cells ing indices of metabolic syndrome tracked with increasing
NO nitric oxide heart failure risk, with two thirds of these patients developing
Nrf2 nuclear factor erythroid 2–related factor 2 heart failure with normal ejection fraction.15 The contempo-
O-GlcNAc O-linked N-acetylglucosamine rary increase in dietary refined carbohydrate, and especially
PI3K phosphatidylinositol 3-kinase fructose, consumption may also impact development of dia-
PKC protein kinase C betic cardiomyopathy as described in more in detail in the
Downloaded from http://ahajournals.org by on February 10, 2022

PPAR peroxisome proliferator-activated receptor molecular mechanisms component of this review.16


RAAS renin–angiotensin–aldosterone system
RAGE receptor for advanced glycation end products
ROS reactive oxygen species
SGLT sodium–glucose cotransporter
SNS sympathetic nervous system

Study found that the incidence of heart failure was increased


in both male and female diabetic patients when compared
with age-matched individuals, and this association was inde-
pendent of obesity, hypertension, dyslipidemia, and coronary
heart disease.2 One study found that the incidence of heart
failure was higher in diabetic (39%) compared with nondia-
betic (23%) patients, with a relative risk of 1.3 for develop-
ing heart failure after 43 months of observation.8 Further data
derived from a population-based observational study in the
Cardiovascular Health Study, the Strong Heart Stud,9 and the
Multi-Ethnic Study of Atherosclerosis10 demonstrated differ-
ences in LV mass and wall thickness and increased diastolic
and systolic dysfunctions between diabetic patients and nor-
Figure1. Pathophysiological mechanisms of diabetic
mal individuals. Meanwhile, in type 1 diabetes mellitus, each cardiomyopathy. Hyperglycemia, insulin resistance, and
1% increase in glycated hemoglobin A1c was linked to a 30% hyperinsulinemia induce cardiac insulin resistance and metabolic
increase for risk of heart failure11 whereas in type 2 diabetes disorders that increase mitochondria dysfunction, oxidative
stress, advanced glycation end products (AGEs), impairment
mellitus, each 1% rise in hemoglobin A1c levels was associ- of mitochondria Ca2+ handling, inflammation, activation of
ated with an 8% increase in risk, independent of other risk renin–angiotensin–aldosterone system (RAAS), autonomic
factors, including obesity, smoking, hypertension, dyslipid- neuropathy, endoplasmic reticulum stress, cardiomyocyte death,
emia, and coronary heart disease,12 suggesting that graded in- as well as microvascular dysfunction. These pathophysiological
abnormalities promote cardiac stiffness, hypertrophy, and
creases in glycemia are a powerful promoter of heart failure fibrosis, resulting in cardiac diastolic dysfunction, systolic
in diabetic patients. dysfunction, and heart failure.
626  Circulation Research  February 16, 2018

Figure 2. The molecular proteins and signaling pathways in hyperglycemia- and insulin resistance-diabetic cardiomyopathy.
Increased protein kinase C (PKC), mitogen-activated protein kinase (MAPK), nuclear factor κ-light-chain-enhancer of activated B cells
(NF-κB), sodium–glucose cotransporter-2 (SGLT2), O-linked N-acetylglucosamine (O-GlcNAc), and cyclic adenosine 5’-monophosphate-
responsive element modulator (CREM) signaling, dysregulation of microRNA (miRNA) and exosomes, and reduction of AMP-activated
protein kinase (AMPK), peroxisome proliferator-activated receptor (PPAR)-γ, and nuclear factor erythroid 2–related factor 2 (Nrf2) induce
cardiac insulin resistance, subcellular component abnormalities, metabolic disorders, and structural changes, resulting in diabetic
cardiomyopathy.

Evolution of Diabetic Cardiomyopathy to Clinical patients, metformin use was associated with a low risk of
Heart Failure mortality in diabetic individuals with heart failure.20 Sodium–
Diabetic cardiomyopathy is usually asymptomatic in the early glucose cotransporter (SGLT) inhibitors and glucagon-like
Downloaded from http://ahajournals.org by on February 10, 2022

stages of its evolution.13 One of the earliest manifestations is peptide 1 receptor agonists have beneficial effects on CVD
LV hypertrophy and decreased LV compliance characterized by outcomes in type 2 diabetic patients.13 A meta-analysis of
impaired early diastolic filling, increased atrial filling, and pro- randomized controlled trials found that dipeptidyl peptidase
longed isovolumetric relaxation.13 LV dilation and symptomatic 4 inhibitors and peroxisome proliferator-activated receptor
heart failure occur after the development of systolic dysfunc- (PPAR) agonists increased the risk of heart failure in patients
tion.13 Indeed, our recent data support the notion that diastolic with or at risk for type 2 diabetes mellitus. The EMPA-REG
dysfunction, as observed by cine magnetic resonance imaging OUTCOME trial (The Empagliflozin Cardiovascular Outcome
in rodents, is associated with impaired cardiac insulin metabolic Event Trial in Type 2 Diabetes Mellitus Patients–Removing
signaling.17 Cardiomyocyte stiffness and hypertrophy, as well Excess Glucose) showed that SGLT2 antagonist treatment
as myocardial fibrosis, all contribute to this cardiac abnormality with empagliflozin reduced primary outcomes, such as non-
(Figure 1). The Cardiovascular Health Study found that, in a fatal myocardial infarction, nonfatal stroke, and CVD-related
cohort of 5201 men and women, the ventricular septal and left mortality, in 7020 diabetic patients.21 Finally, treatment with
posterior myocardial wall thicknesses were greater in diabetic 2 long acting glucagon-like peptide 1 receptor agonists sig-
patients than in nondiabetic individuals and that this was associ- nificantly reduced CVD events and heart failure in high-risk
ated with compromised systolic or diastolic function.18 diabetic patients.22,23 The results suggest that these antidiabetic
There is a considerable body of epidemiological evidence drugs may have a role in preventing and treating diabetic car-
that implicates obesity, linked to increased intake of refined diomyopathy and associated CVD in type 2 diabetic patients.
carbohydrates and decreased exercise, in the increasing preva-
lence of diabetes mellitus and related heart disease throughout Functional Phenotype of Diabetic
the world. Lifestyle changes such as aerobic exercise, weight Cardiomyopathy
control, and smoking cessation are efficacious therapeutic The first stage of diabetic cardiomyopathy is clinically as-
approaches in the prevention of diabetic cardiomyopathy. ymptomatic and is characterized by increased fibrosis and
Sustained glycemic control reduces the prevalence of diabetic stiffness; there is a reduction of early diastolic filling and an
cardiomyopathy and reduces cardiovascular disease (CVD). increase in atrial filling and enlargement, as well as an el-
For example, normalization of glycemia with insulin therapy evated LV end-diastolic pressure.24 Underlying pathological
reduced cardiomyocyte hypertrophy, collagen content, and factors include hyperglycemia, systemic and cardiac insulin
diastolic dysfunction and limited progression of diabetic resistance, increased free fatty acid (FFA) levels, systemic
cardiomyopathy in type 1 diabetic rodent models.19 There is and tissue inflammation, oxidative stress, and activation of the
emerging evidence that some glycemic therapies may have renin–angiotensin–aldosterone system (RAAS) and the sym-
specific benefits. In a retrospective cohort study of 10 920 pathetic nervous system (SNS; Figure 1).13 Reduced calcium
Jia et al   Diabetic Cardiomyopathy and Heart Failure   627

(Ca2+) pump activity-induced inefficient sequestration of sar- decreases glucose transporter type 4 (GLUT4) recruitment
coplasmic reticulum Ca2+ is regarded as an important contrib- to the plasma membrane and glucose uptake, thus lowering
utor to the development of the cardiac diastolic dysfunction.25 sarcoplasmic reticulum Ca2+ pump activity and increasing car-
The second stage of diabetic cardiomyopathy is charac- diomyocyte intracellular Ca2+.13 Meanwhile, abnormal insulin
terized by LV hypertrophy, cardiac remodeling, advancing metabolic signaling also decreases insulin-stimulated coronary
cardiac diastolic dysfunction, and the consequent emergence endothelial nitric oxide (NO) synthase (eNOS) activity and
of clinical indications of heart failure with normal ejection NO production increasing cardiomyocyte intracellular Ca2+/
fraction.13 With progression of diabetic cardiomyopathy, dia- Ca2+ sensitization and reducing sarcoplasmic Ca2+uptake.13
stolic dysfunction and reduced cardiac compliance may co- Reduction of NO bioavailability may also lead to phosphory-
exist with systolic dysfunction leading to reduced ejection lation of titin increasing the ratio of stiff titin isoform N2B/
fraction, prolonged pre-ejection performance, an enlarged LV N2BA (compliant) expression. These pathophysiological ab-
chamber, shortened ejection period, and the latter by an in- normalities increase cardiac stiffness and impair relaxation,
creased resistance to filling with increased filling pressures.13 cardinal manifestations of diabetic cardiomyopathy.13 Other
Abnormalities in contractile and regulatory protein expression pertinent abnormalities include hyperglycemia, insulin resis-
are responsible for the mechanical defects in cardiac contrac- tance, and oxidative stress that promote expression of several
tion. For example, decreased Ca2+ sensitivity along with shifts cardiomyocyte hypertrophic genes, such as β-myosin heavy
in cardiac myosin heavy chain from V1 to V3 isoforms con- chain, insulin-like growth factor 1 receptor, and B-type na-
tributes to the impaired cardiac systolic dysfunction, a long- triuretic peptide.31 High insulin levels induce cardiomyocyte
term complication of diabetes mellitus.26 Phosphorylation of hypertrophy by binding to the insulin-like growth factor 1
troponin also contributes to depressed myocardial contractil- receptor. Insulin-like growth factor 1, produced by cardio-
ity because myosin light chain-2 and troponin I are involved myocytes, can also stimulate cardiomyocyte hypertrophy via
in regulating cardiomyocyte contraction.27 the insulin receptor, extracellular signal-regulated kinase 1/2
The phenotypes and underlying mechanisms of diabetic (Erk1/2), and phosphatidylinositol 3-kinase (PI3K) signaling
cardiomyopathy in type 2 diabetes mellitus have been mostly pathways.32 Crosstalk between the insulin-like growth factor 1
investigated in db/db mice, ob/ob mice, Zucker diabetic fat- and insulin signaling pathways plays an important role in hy-
ty rats, and diabetic patients.13 The impact of type 1 diabe- perglycemia/insulin resistance–induced cardiac hypertrophy
tes mellitus on systolic and diastolic functions is less clear. and fibrosis in diabetic cardiomyopathy, as described in more
As in type 2 diabetes mellitus, diastolic dysfunction is also detail later in this review.
often observed in type 1 diabetes mellitus.28 The underlying Development of myocardial fibrosis in diabetic cardiomy-
Downloaded from http://ahajournals.org by on February 10, 2022

mechanisms of diabetic cardiomyopathy in type 1 diabetes opathy involves the deposition of stiff collagen and its cross-
mellitus probably mostly overlap, but different alterations ex- linking, cardiac interstitial fibrosis, progressive abolition of
ist in hearts of type 2 diabetes mellitus.29 For instance, sys- muscular fibrils, perivascular fibrosis, thickened and sclerotic
tolic function was preserved, and cardiac hypertrophy was not small coronary vessels, and basement membrane thickening,
observed in type 1 Akita diabetic mice although hearts were as well as coronary microvascular sclerosis and microaneu-
smaller compared with nondiabetic controls.30 Cardiomyocyte rysms.33,34 Activation of the RAAS and SNS, stimulation of
autophagy was enhanced in type 1 but suppressed in type 2 advanced glycation end products (AGE)–mediated signaling
diabetes mellitus.28 Clearly, further studies are necessary to via cell surface receptor for AGE (RAGE), hyperinsulinemia,
understand the potential differences in phenotype and under- and hyperglycemia collectively lead to activation of trans-
lying mechanisms for diabetic cardiomyopathy in type 1 and forming growth factor β1 pathway and dysregulation of ex-
type 2 diabetes mellitus. tracellular matrix degradation.35 Some biomarkers of collagen
Thus, cardiac dysfunction in diabetic hearts progresses synthesis, including inflammation cytokines, connective tis-
from subclinical cardiac abnormalities, such as LV fibrosis, sue growth factor, metalloproteinases, and galectin-3, can be
to diastolic dysfunction and eventually systolic dysfunction used clinically in the determination of myocardial fibrosis.36
accompanied by reduced ejection fraction. Several nonin- As discussed in more detail later, reduced bioavailable NO, in-
vasive techniques, including echocardiography, computed creased oxidative stress, and an activated transforming growth
tomography, and cinematic magnetic resonance imaging, factor β1/SMAD signaling pathway, in concert with impaired
have been applied to detect changes of cardiac structure (ie, insulin metabolic signaling pathways, increase the myocardial
fibrosis) and function.13 Furthermore, elevated levels of atrial fibronectin and collagen content and cardiac interstitial fibro-
natriuretic peptide, brain natriuretic peptide, and O-linked sis characteristic of diabetic cardiomyopathy.17
N-acetylglucosamine (O-GlcNAc), among others, may also
Cardiac Insulin Resistance and Diabetic
serve as markers for diabetic cardiomyopathy and heart
Cardiomyopathy
failure.13
Cardiac insulin signaling mediates cellular homeostasis via
control of protein synthesis, substrate use, and cell survival.
Molecular Mechanisms Underlying Diabetic Glucose transport in cardiac tissue is mediated via GLUT4 as
Cardiomyopathy in skeletal muscle, liver, and fat tissue. Insulin, binding to the
Cardiac Structural Abnormalities insulin receptor, activates insulin signaling/docking molecule
The mechanism promoting cardiomyocyte stiffness in the dia- insulin receptor substrate (IRS)-1/2 and downstream PI3K/
betic heart include impaired insulin metabolic signaling that protein kinase B (Akt), stimulating GLUT4 translocation to the
628  Circulation Research  February 16, 2018

cell membrane and subsequent glucose uptake.13 Furthermore, be isomerized into glyceraldehyde 3-phosphate and acetyl-
normal coronary artery and myocardial insulin metabolic sig- coenzyme A (CoA). Acetyl-CoA is either oxidized in the tri-
naling promotes eNOS activation and bioavailable NO neces- carboxylic acid cycle or directed toward FFA synthesis.49,50
sary for optimal coronary microvascular flow and myocardial Phosphorylation of fructose also decreases ATP produc-
function.13,17 Cardiac insulin receptor knockout decreases car- tion.49,50 Enhanced cellular fructose metabolism promotes sev-
diac glucose uptake, increases cardiac reactive oxygen species eral subcellular hexose sugar-related protein modifications,
(ROS) production, and induces mitochondrial dysfunction.37,38 such as O-GlcNAc, and formation of AGE that contributes to
Double IRS-1/2 knockout reduces cardiomyocyte ATP con- impaired insulin metabolic signaling, reductions in NO pro-
tent, impairs cardiac metabolism and function, and increases duction, and increased cardiac fibrosis.51
fibrosis and cardiac failure.37,38 Reduced PI3K/Akt signaling
and reduced GLUT4 expression and translocation have also Decreased Flexibility in Substrate Using in Diabetic
been found in ventricular muscle biopsies obtained from pa- Cardiomyopathy
tients with type 2 diabetes mellitus.39 The E3 ubiquitin ligase, Under normal physiological circumstances, the heart displays
mitsugumin 53, may play an important negative role in the considerable metabolic substrate flexibility, using energy from
maintenance of insulin signaling.40 Elevated cardiac mitsugu- various substrates, such as FFAs, glucose, ketone bodies,
min 53 protein levels in a type 2 diabetic mouse model were lactate, and some amino acids to produce ATP, the predomi-
correlated with increased proteosomal degradation of the insu- nant source of energy for cardiac energetics.13 Mitochondria
lin receptor and IRS-1. Furthermore, cardiomyocyte-specific normally occupy ≈20% to 30% of the total cell volume of
overexpression of mitsugumin 53 inhibited insulin signaling cardiomyocytes.13 Typically, mitochondrial oxidative phos-
and increased cardiac fibrosis,41 suggesting that downregula- phorylation produces >95% of ATP.52 The citric acid cycle
tion of cardiac mitsugumin 53 may be a potential therapeutic usually accounts for the remaining 5% of ATP produced from
strategy in the prevention of diabetic cardiomyopathy and pro- glucose and lactate in the heart.53 However, in the setting of
gression to clinically manifested heart failure. hyperglycemia, insulin resistance, and hypertriglyceridemia,
Risk factors, such as obesity and inappropriate activa- there is a reduction in the myocardium’s ability to use glucose
tion of RAAS, can impair cardiac insulin metabolic signal- as an energy source, and it subsequently switches to FFAs.53
ing through enhanced activation of the mammalian target This energy substrate switch is accompanied by impaired oxi-
of rapamycin (mTOR)/S6 kinase 1 signaling pathway,13,17 dative phosphorylation and a mitochondrial proton leak that
which increases serine phosphorylation and reduces tyrosine results in increased production of ROS. As the heart has lim-
phosphorylation of IRS-1/2 and impairs PI3K engagement ited antioxidant capacity, increased mitochondrial ROS pro-
Downloaded from http://ahajournals.org by on February 10, 2022

and Akt/eNOS activation and NO production.13,17 Lower NO duction leads to NO destruction and reduced bioavailable NO,
production impairs coronary vessel relaxation and insulin- hallmarks of diabetic cardiomyopathy.1,22
mediated capillary recruitment, both of which are important
Role of Abnormal FFA Metabolism in Diabetic
for the delivery of insulin and glucose necessary for normal
Cardiomyopathy
myocardial energetics.42–45 Impairment of NO production also
Increased FFA release from adipose tissue and increased ca-
leads to increased activation of collagen cross-linking en-
pacity of myocyte sarcolemmal FFA transporters also contrib-
zymes, such as transglutaminase, thereby promoting cardiac
ute to the development of diabetic cardiomyopathy.13,53 Cluster
fibrosis and stiffness.1,10 Proinflammatory cytokines, such as
of differentiation 36 (CD36), a predominantly membrane-
tumor necrosis factor α , have been reported to induce car-
located protein and transporter that promotes FFA uptake in
diac insulin resistance through activation of nuclear factor
both sarcolemma and endosomal membranes, is increased in
κ-light-chain-enhancer of activated B cells (NF-κB) and c-
diabetic hearts.54 Increased subcellular vesicular recycling of
Jun N terminal kinase (JNK) that induce phosphorylation of
CD36 from endosomes to the plasma membrane increases
IRS-1.13 Activation of forkhead box–containing protein 1, O
the rate of cellular FFA uptake in diabetic hearts.13,54 CD36
subfamily, directly regulates IRS-1 signaling and decreases
is also paramount in AMP-activated protein kinase (AMPK)–
PI3K/Akt signaling, leading to insulin resistance in mice fed a
mediated stimulation of FFA uptake in cardiomyocytes.
high-fat diet.46 Furthermore, deletion of cardiac forkhead box–
Indeed, CD36 knockout mice show a 70% reduction in FFA
containing protein 1, O subfamily, largely prevented heart
uptake in cardiomyocytes,55 and CD36 deficiency rescues li-
failure in these animals.47 Thus, forkhead box–containing
potoxic cardiomyopathy.56,57 Activation of AMPK is respon-
protein 1, O subfamily, may also provide a novel therapeu-
sible for early activation of glucose uptake and glycolysis and
tic or preventive strategy for treating individuals with diabetic
improves cardiac function in diabetic patients.58 However,
cardiomyopathy.47
AMPK activation is attenuated in diabetes mellitus increasing
Role of Fructose in the Pathogenesis of Diabetic FFA uptake and triacylglycerol accumulation and reducing
Cardiomyopathy glucose use, also characteristic of diabetic cardiomyopathy.59
High-fructose diets induce cardiomyocyte autophagy, oxida- Several lipid metabolites, such as diacylglycerols and ce-
tive stress, and impaired insulin metabolic PI3K/Akt/eNOS ramides, impair insulin metabolic signaling contributing to and
signaling and interstitial fibrosis.16,48 Generally, fructose is exacerbating diabetic cardiomyopathy. Insulin sensitivity in
readily absorbed and rapidly metabolized by the human liver obese humans can be correlated to elevated diacylglycerol con-
through GLUT2 and 5.13 Fructose 1-phosphate is cleaved to tent and PKC ε (protein kinase Cε) activation.60 Diacylglycerol
dihydroxyacetone phosphate by aldolase B, which can then increases lipid-associated endoplasmic reticulum stress.60 A
Jia et al   Diabetic Cardiomyopathy and Heart Failure   629

high-fat diet increases diacylglycerol in the membrane fraction, in type 2 diabetes mellitus, mitochondria switch from glucose
activating PKCε and inducing insulin resistance,61 and lower- to FFA oxidation for ATP production (Figure 1).13 This is ac-
ing NO production.62 Comparative gene identification 58 is a companied by increased mitochondrial ROS generation and
lipid droplet-associated protein that promotes triglyceride hy- impaired oxidative phosphorylation. Altered mitochondrial
drolysis and adipose triglyceride lipase activation.63 Inhibition Ca2+ handling further promotes mitochondrial respiratory dys-
of comparative gene identification 58 induced hepatic steatosis function leading to cell death.72 Metabolic stress–induced mi-
and increased total diacylglycerol content in the absence of tochondrial dysfunction also increases Ca2+ overload–induced
hepatic insulin resistance.63 Studies have found that compara- opening of the mitochondrial permeability transition pores
tive gene identification 58 gene knockout prevents diacylglyc- resulting in cardiomyocyte autophagy and cardiac necrosis.73
erol accumulation at the plasma membrane, PKCε activation,
and the impairment of hepatic insulin metabolic signaling.61,63 Mitochondrial Oxidative Stress in the Pathogenesis
Meanwhile, ceramide can directly activate atypical PKCs and of Diabetic Cardiomyopathy
inhibit insulin metabolic Akt/PKB (protein kinase B) signal- Oxidative stress promotes development and progression of
ing, attenuating GLUT4 translocation and insulin-stimulated cardiac insulin resistance, diabetic cardiomyopathy, and heart
glucose uptake in diabetic hearts.62 These data support a role failure (Figure 1). Mitochondrial ROS are a natural byproduct
for intracellular compartmentation of diacylglycerol and ce- of oxygen metabolism at complexes I and III within the elec-
ramide in causing lipotoxicity and metabolic insulin resistance tron transport chain.13 Under normal physiological conditions,
in several tissues, including the heart. the major electrochemical proton gradient is used to synthe-
tize ATP.13 However, hyperglycemia and insulin resistance
Abnormalities in Ketogenesis in Diabetic increase the nicotinamide adenine dinucleotide and flavin ad-
Cardiomyopathy enine dinucleotide flux to the mitochondrial respiratory chain,
Type 2 diabetes mellitus is often associated with decreased resulting in hyperpolarization of the mitochondrial inner
ketogenesis because of systemic insulin resistance and hyper- membrane, inhibition of electron transport in complex III, and
insulinemia.64 Ketones, such as B-hydroxybutyrate, may play excess ROS production.74 Nicotinamide adenine dinucleotide
a key role in maintaining bioenergetic homeostasis in diabetic phosphate oxidase is another important source of cardiomyo-
cardiomyopathy, where there is reduced cardiac glucose use.65 cyte ROS. Increased cardiomyocyte nicotinamide adenine
In this regard, treatment with empagliflozin, an SGLT2 an- dinucleotide phosphate oxidase activity has been reported
tagonist, increased ketone levels providing a more efficient in diet-induced obesity and systemic and cardiac insulin re-
energy source in the failing myocardium of diabetic patients sistance.75 Increased RAAS-mediated nicotinamide adenine
with heart failure,66 perhaps compensating for an impairment
Downloaded from http://ahajournals.org by on February 10, 2022

dinucleotide phosphate oxidase activity may also directly pro-


in mitochondrial energy transduction related to decreased mote cardiac fibrosis by activation of a profibrotic transform-
myocardial glucose use. ing growth factor β1/Smad 2/3 signaling pathway.76,77 Other
Abnormalities in Cardiac Glucose FFA Cycling sources of ROS in diabetic cardiomyopathy include increases
The glucose fatty acid cycle (Randle cycle) is a metabolic in xanthine oxidase, and microsomal P-450 enzyme activity,
mechanism that involves competition between glucose and and uncoupling of NO synthase. Elevated cardiac tissue ROS
FFAs for their oxidation and uptake and is thus related to in- not only increases polyol pathway flux, formation of AGEs,
sulin resistance and type 2 diabetes mellitus. In this regard, ac- expression of the receptor for AGEs and its activating ligands,
tivation of lipolysis provides FFAs as the preferred fuel source PKC signaling, and the hexosamine pathway but also inhibits
leading to ketogenesis during fasting whereas inhibition of eNOS and prostacyclin synthase activity, all of which con-
glucose oxidation contributes to a glucose-sparing effect that tribute to the development of cardiomyopathy as previously
is an essential survival mechanism during times of starva- reviewed.78
tion.67 In the fed state or during exercise, glucose is rerout-
Role of AGEs and RAGE in the Pathophysiology of
ed to glycogen, and muscle glycogen content is increased.67
Diabetic Cardiomyopathy
Typically, FFA oxidation increases the mitochondrial ratios
Hyperglycemia increases AGE accumulation and induces
of acetyl-CoA/CoA and nicotinamide adenine dinucleotide/
myocardial structural alterations by increases in nonenzy-
NAD+ that inhibit pyruvate dehydrogenase activity and im-
matic glycation, oxidation of lipids and proteins, myocardial
pair glucose metabolism, resulting in accumulation of cyto-
collagen and fibronectin production, and connective tissue
solic citrate, increased glucose 6-phosphate, and inhibition of
cross-linking and fibrosis (Figure 1).13 Indeed, AGE-induced
hexokinase.67,68 Meanwhile, glucose oxidation produces ci-
connective tissue cross-linking of extracellular matrix pro-
trate that can be metabolized to malonyl-CoA; malonyl-CoA
motes myocardial fibrosis and impaired passive relaxation.13
controls the entry and oxidation of long chain FFA favoring
AGEs may also bind to RAGE, which further promotes mal-
FFA esterification.67,69,70
adaptive structural changes and impaired myocardial ener-
Mitochondrial Dysfunction in the Genesis of getics.13 For example, interaction of AGEs with RAGE on
Diabetic Cardiomyopathy cardiomyocyte surfaces induces maladaptive proinflamma-
Mitochondrial dysfunction plays a pivotal role in the devel- tory responses and increases matrix protein and connective
opment of diabetic cardiomyopathy and associated heart tissue production, mediated through Janus kinase and mito-
failure.71 Mitochondrial oxidative phosphorylation provides gen-activated protein kinase (MAPK) pathway activation.13
90% of intracellular ATP production in cardiomyocytes, but In addition, AGEs elevation increases ROS production and
630  Circulation Research  February 16, 2018

transforming growth factor β1/SMAD pathway activation and pathways involving NF-κB, chemokines, and ROS. An NF-
connective tissue production and fibrosis. In a prospective κB–positive feedback loop further increases NLRP3 in-
clinical study of 194 patients with acute coronary syndrome, flammasome assembly and procaspase-1 activation and
AGEs were an independent risk factor in postinfarction heart pro-interleukin-1β processing and maturation.88 Meanwhile,
failure.79 Furthermore, increases in plasma AGEs indepen- increased monocyte/macrophage migration through the coro-
dently predicted mortality and hospitalization for heart failure nary endothelium increases resident cardiac macrophages,
in a study of 580 diabetic patients.80 which can be polarized into proinflammatory M1 phenotypes
under conditions of increased ROS and reduced bioavailable
Impaired Mitochondrial Ca2+ Handling in Diabetic NO.13,17 In recent investigative work, it has been observed that
Cardiomyopathy macrophage proinflammatory M1 polarization is upregulated
Cytosolic Ca2+ levels regulate cellular metabolism, muscle whereas macrophage M2 anti-inflammatory response is re-
contraction, and cell signaling (Figure 1). Typically, in cardiac pressed in diabetic heart tissues.13,17
excitation–contraction coupling, Ca2+ enters the cytoplasm
through voltage-sensitive L-type Ca2+ channels after depolar- Activated RAAS in the Genesis of Diabetic
ization of the sarcolemma, and this triggers Ca2+ release from Cardiomyopathy
the sarcoplasmic reticulum. The Ca2+ binds troponin C to in- Increased activation of the systemic and tissue RAAS in states
duce myofibrillar contraction.81,82 During cardiac relaxation, of hyperglycemia and insulin resistance plays an important
Ca2+ is transported back into the sarcoplasmic reticulum, and role in the pathogenesis of diabetic cardiomyopathy and heart
the remaining cardiomyocyte Ca2+ is pumped out by the sar- failure (Figure 1). Serum angiotensin II levels are significantly
colemma Na+/Ca2+ exchanger and the plasma membrane Ca2+ correlated with postprandial glucose concentrations in insu-
pump.81,82 However, in diabetic cardiomyopathy, impaired lin resistance and type 2 diabetes mellitus.89 Furthermore, the
Ca2+ handing by all of these transporters increases action proinflammatory angiotensin II receptor 1 is upregulated and
potential duration and prolongs diastolic relaxation time.13 the anti-inflammatory AT-2R is downregulated in early diabe-
Elevated intracellular resting Ca2+, prolongation of intracellu- tes mellitus.90 Exercise induces a shift of the RAAS toward the
lar Ca2+ decay, slowed Ca2+ transients, reduction of sarcoplas- angiotensin-converting enzyme 2/Mas receptor axis in skel-
mic reticulum Ca2+ pumping, and impairment of sarcoplasmic etal muscle, providing protection in obese rats.91 Conversely,
reticulum Ca2+ reuptake have been documented in hearts of inhibition of the AT2 receptor with PD123319 impairs insulin
type 2 diabetic mice.83,84 Similar changes are seen in type 1 signaling in C57BL/6 mice.92 In patients with type 1 diabe-
diabetic rodent models.85,86 The data suggest that impaired car- tes mellitus, hyperglycemia-induced activation of systemic
Downloaded from http://ahajournals.org by on February 10, 2022

diomyocyte Ca2+ handling plays a key role in the development RAAS seems to play a role in the pathogenesis of diabetic car-
of the cardiac diastolic dysfunction characteristic of early dia- diomyopathy.93,94 Experimental evidence also supports a role
betic cardiomyopathy. for increased mineralocorticoids in systemic and tissue insu-
lin resistance.17 High plasma aldosterone and overexpression
Inflammation as an Instigator of Diabetic of the tissue mineralocorticoid receptors (MR) are associated
Cardiomyopathy with systemic insulin resistance, hyperglycemia, and dyslip-
A maladaptive proinflammatory response has been impli- idemia.95 Large randomized controlled trials have shown that
cated in the development of diabetic cardiomyopathy. The in- inhibition of the aldosterone/MR signaling pathway reduces
nate immune system, that is, neutrophils, mast cells, dendritic morbidity and mortality in diabetic patients with both mild and
cells, macrophages, and eosinophils, is involved (Figure 1).13 moderately severe heart failure.62 RAAS activation impairs in-
Activation and expression of proinflammatory cytokines, such sulin metabolic signaling and induces systemic and cardiac
as tumor necrosis factor α, interleukins 6 and 8, monocyte insulin resistance, in part, through activation of the mTOR/S6
chemotactic protein 1, adhesion molecule intercellular adhe- kinase 1 signaling pathway.96 Meanwhile, enhanced angioten-
sion molecule 1, and vascular cell adhesion molecule 1, all sin II receptor 1 and MR activation increases coronary artery
contribute to cardiac oxidative stress, remodeling and fibrosis, endothelial leukocyte/monocyte adhesion, proinflammatory
and diastolic dysfunction.13 Cytokine expression is regulated by cytokine expression, macrophage infiltration, and polarization
the nuclear transcription factor, NF-κB.13 Finally, the Toll-like resulting in an increase in the proinflammatory M1 phenotype
receptor-4 also plays an important role in triggering increased in the myocardium. These abnormalities exacerbate the mal-
NF-κB, proinflammatory and innate immune system respons- adaptive cardiac remodeling, interstitial fibrosis, and diastolic
es.87 These proinflammatory responses occur in different pop- dysfunction seen in diabetic cardiomyopathy.17
ulations of cardiac cells, including coronary endothelial and
smooth muscle cells, as well as fibroblasts and cardiomyocytes. Autonomic Neuropathy in Diabetic
High FFA levels, impaired insulin metabolic signaling, and Cardiomyopathy
hyperglycemia activate the NLRP3 (NACHT, LRR, and PYD Cardiac autonomic neuropathy is a secondary complication
domains-containing protein 3) inflammasome, a novel mo- related to sustained hyperglycemia (Figure 1) and includes ab-
lecular marker in diabetic cardiomyopathy.88 Separation from normalities in heart rate control, vascular hemodynamics, and
cytoplasmic chaperones and oligomerization follows NLRP3 cardiac structure and function.13,97,98 An early characteristic
activation, leading to recruitment of procaspase-1.88 Activated of cardiac autonomic neuropathy is reduction of parasympa-
caspase-1 processes interleukin-1β and interleukin-18 precur- thetic activity with an imbalance toward relatively higher SNS
sors and serves as an enhancer of multiple proinflammatory activity.99,100,101 In this regard, activation of the SNS enhances
Jia et al   Diabetic Cardiomyopathy and Heart Failure   631

β-1 adrenergic receptor (β1) signaling that promotes cardiac MR antagonist improved coronary microvascular function and
hypertrophy, interstitial fibrosis, cardiomyocyte apoptosis, prevented CVD in patients with type 2 diabetes mellitus, sug-
and impaired function.98 gesting an important role of cardiac MR activation in coronary
microvascular dysfunction in diabetes mellitus.106 Structural
Endoplasmic Reticulum Stress and Increased Cell abnormalities in the coronary microcirculation include lumi-
Death in Diabetic Cardiomyopathy nal obstruction, inflammation infiltration, vascular remodel-
Cardiac oxidative stress, lipotoxicity, inflammation, and the ing, and perivascular fibrosis.107 Functional abnormalities in
accumulation of misfolded proteins impair the function of the coronary microcirculation include endothelial and smooth
cardiac endoplasmic reticulum and promote endoplasmic muscle cell dysfunction and impairment of vascular relax-
reticulum stress, inducing the unfolded protein response ation and constriction and ischemic reperfusion.107 The normal
(Figure 1).13 Together, endoplasmic reticulum stress and the function of coronary arteries, and downstream microcirculato-
unfolded protein response inhibit cellular protein synthesis ry vessels, is impaired in diabetic cardiomyopathy (Figure 1).
and degradation of misfolded or damaged proteins and ul- Physiologically, several vasoactive substances, including NO,
timately increase cell apoptosis and autophagy.13 Increased prostacyclin, and endothelium-derived hyperpolarizing fac-
cardiac apoptosis is a major risk factor for the development tors, released from endothelial cells lining the coronary mi-
of diabetic cardiomyopathy; biopsied diabetic heart tis- crocirculation, play a beneficial vasodilatory role.42 Indeed,
sue expresses 85-fold more cardiomyocyte apoptosis than although NO-mediated vasodilation may be impaired, vascu-
control nondiabetic hearts.101 Endoplasmic reticulum stress lar function in the early stages of diabetes mellitus is often pre-
also induces autophagy through a Ca2+-dependent pathway, served through normal or even enhanced endothelium-derived
involving the inositol-requiring enzyme 1 and protein ki- hyperpolarizing factor–induced vasodilation.42 However, both
nase RNA-like endoplasmic reticulum kinase pathways.102 NO- and endothelium-derived hyperpolarizing factor–induced
Typically, autophagy is regulated through the mTOR, AMPK, vasodilation are eventually affected, leading to significant
and silent information regulator pathways.13 mTORC1 has dysfunction of the microcirculation in the later stages of dia-
been proposed to regulate autophagy by repressing the au- betes mellitus.42 Recent investigation has also suggested that
tophagy-related 1 (Atg1)–Atg13–Atg101/FIP200 (FAK fam- persistently high plasma endothelin-1 levels, along with re-
ily–interacting protein of 200 kDa) complex. Thus, inhibition duced eNOS activity and NO production, are associated with
of mTORC1 facilitates the initiation of autophagy.102 The the development of cardiac fibrosis and diastolic dysfunction
inositol-requiring enzyme 1 arm of endoplasmic reticulum in diabetic patients.108 In this regard, endothelial cell–specific
stress leads to JNK activation and increased phosphorylation endothelin-1 knockout prevented diabetes mellitus–induced
Downloaded from http://ahajournals.org by on February 10, 2022

of Bcl-2, which promotes its dissociation from Beclin-1.102 cardiac fibrosis and exerted a beneficial effect in the preven-
Thus, enhanced mTOR may serve as a convergence point for tion of diabetic cardiomyopathy.108
abnormalities involving the interplay between endoplasmic
reticulum stress and autophagy in the hearts of diabetic indi- Molecular Protein Signature in Diabetic
viduals. However, in diabetic cardiomyopathy, dysregulation Cardiomyopathy
of autophagy impairs cardiomyocyte autophagosome and Several molecular proteins and signaling pathways have been
lysosome fusion.103 One study found that chronic metformin implicated as important contributors to the development of
treatment activated AMPK activity, restored autophagic ac- diabetic cardiomyopathy and heart failure. These include
tivity, and inhibited cardiomyocyte apoptosis by disrupting AMPK, PPARs, O-GlcNAc, SGLT2, PKC, MAPK, NFκB,
the Bcl-2 and Beclin-1 complex in diabetic heart tissue.103 nuclear factor erythroid 2–related factor 2 (Nrf2), cyclic ad-
Therefore, the activation of the autophagic response is usu- enosine 5′-monophosphate-responsive element modulator
ally regarded as a compensatory feedback mechanism to (CREM), microRNAs (miRNA), and exosomes as discussed
protect against cell apoptosis and to maintain normal cellular below (Figure 2).
function in conditions, such as insulin resistance and type 2
diabetes mellitus. Impaired AMPK Activation in Diabetic
Cardiomyopathy
Microvascular Dysfunction in Diabetic AMPK is a master regulator of cellular energy homeosta-
Cardiomyopathy sis.13 With cellular stress and an increased AMP/ATP ratio,
Diabetic cardiomyopathy is classically defined in the context AMPK activation enhances expression and translocation
of absence of overt coronary artery disease. Nevertheless, dia- of GLUT4 and thus insulin-induced glucose uptake and in-
betic cardiomyopathy may be associated with coronary mi- creases mitochondrial biogenesis leading to FFA oxidation
crovascular dysfunction that impairs coronary blood flow and and glycolysis.58 Specifically, in cardiomyocytes, activated
myocardial perfusion, ventricular function, and clinical out- AMPK positively stimulates glucose uptake, FFA oxidation,
comes, including CVD.104 In a clinical study of 2783 consecu- and glycolysis while negatively regulating mTOR signaling,
tive patients, impaired coronary flow reserve was associated gluconeogenesis, and lipid and protein synthesis.109 Therefore,
with an adjusted 3.2- and 4.9-fold increase in the rate of cardi- AMPK activation plays a beneficial role in preventing the pro-
ac death for both diabetic and nondiabetic individuals, respec- gression of diabetic cardiomyopathy. For this reason, AMPK
tively, indicating that coronary microcirculation dysfunction has been considered as a promising target for drug discovery
is a powerful, independent correlate of cardiac mortality and development for the prevention and reversal of diabetic
among both diabetics and nondiabetics.105 Treatment with an cardiomyopathy.
632  Circulation Research  February 16, 2018

Alterations in Activation of Cardiac PPARs the diabetic heart mediates diabetes mellitus–induced impair-
Several different PPAR isoforms, namely α, β/δ, and γ, are ex- ment of insulin metabolic signaling, cardiomyocyte apoptosis,
pressed in the heart and play a key role in myocardial glucose myocardial excitation–contraction coupling, and cardiac relax-
and lipid metabolism and energy homeostasis. Importantly, ation.119 Overexpression of O-GlcNAcase removes O-GlcNAc
they also exert roles not directly related to metabolism, such and restores normal cardiomyocyte Ca2+ handling and cardiac
as inflammation and oxidative stress.13 PPAR-α is expressed at function,120 suggesting that targeting of hexosamine biosyn-
relatively high levels in the heart, and its activation directly im- thesis and O-GlcNAc may be a fruitful potential strategy for
pacts FFA uptake and mitochondrial FFA oxidation.13 PPAR- the prevention and therapy of diabetic cardiomyopathy.
α regulates lipoprotein assembly and transport and modulates
both oxidant and antioxidant defenses.110 Overexpression of SGLT2 Abnormalities and Potential Cardiac
cardiomyocyte-specific PPARα causes decreased uptake of Benefits of Inhibition of This Transporter
Ca2+ by the sarcoplasmic reticulum, LV hypertrophy, systolic Glucose is actively transported from the gut lumen into the
dysfunction, and increased atrial natriuretic and B-type natri- gastrointestinal epithelium primarily by SGLT1, highly ex-
uretic peptide expression.59 Conversely, deletion of cardiac pressed in the brush border membrane of enterocytes.121,122 In
PPAR-α prevents fasting-induced expression of FFA meta- hyperglycemia and insulin resistance, glucose and fructose
bolic genes and induces a switch from FFA to glucose use.111 absorption through the intestinal mucosa increases because
With the development of diabetic cardiomyopathy, chronic ex- of higher expression of SGLT1, GLUT2, and GLUT5 and in-
posure to elevated FFAs seems to reduce PPAR-α expression. creased brush border disaccharidases, sucrase, maltase, and
In rodent cardiomyocytes, this was shown to further decrease lactase activity.121,122 SGLT2 is exclusively expressed in the
cardiac function by inhibition of FFA oxidation and increased kidney and mostly localized in the brush border membrane
intracellular fat accumulation.112,113 However, human studies of proximal tubule epithelial cells in the S1 segment of the
suggest that PPAR-α expression is not significantly altered proximal convoluted tubule.121,122 SGLT2 expression is sig-
in the hearts of patients with type 2 diabetes mellitus.72,114 nificantly increased in diabetic humans,123 rats,124 and db/db
Activated PPAR-α–induced increases in FFA oxidation and mice,125 and this is correlated with glomerular hyperfiltra-
use in the diabetic heart may thus initially serve as a compen- tion, increased glucose reabsorption, and elevated plasma
satory mechanism to adjust substrate oxidation to available glucose.126 Conversely, SGLT2 inhibition leads to natriure-
substrate supply. Furthermore, reduced PPAR-α in advanced sis, osmotic diuresis, plasma volume contraction, and reduc-
disease may have maladaptive consequences in terms of car- tion of blood pressure and arterial stiffness, all mechanisms
diac metabolism, including glucotoxicity and functional car- that may mitigate diabetic cardiomyopathy and heart failure.
Downloaded from http://ahajournals.org by on February 10, 2022

diac abnormalities. The role of PPAR-α in the development Furthermore, SGL2 inhibitor treatment can shift cell metabo-
of cardiac dysfunction in diabetic cardiomyopathy has been lism from glucose to FA oxidation. Thus, the cardiac benefi-
inadequately evaluated and needs to be further investigated. cial effects of SGLT2 may include making more of the ketone
Similar to PPAR-α, PPAR-β/δ isoforms are also expressed body, B-hydroxybutyrate, a highly energy-efficient substrate
abundantly in heart tissue and regulate transcriptional gene for cardiac metabolism. Targeting SGLT2 has been shown to
expression and FFA metabolism.115 Enhanced PPAR-β/δ sig- improve cardiovascular outcomes and mortality in type 2 dia-
naling promotes FFA use whereas deletion of PPAR-β/δ de- betic patients (Figure 2).21 Ongoing research is addressing the
creases FFA oxidative gene expression and FFA oxidation.115 role of these agents for specific prevention and treatment of
In addition, PPAR-γ plays important roles in cardiac antihy- heart failure in diabetic individuals.127
pertrophic and anti-inflammatory effects.13 PPAR-γ agonists
PKC Activation Promotes Development of Diabetic
enhance cardiomyocyte insulin sensitivity and improve car-
Cardiomyopathy
diomyocyte glucose uptake.13 Thus, PPAR-γ may be benefi-
PKC signaling pathways are activated in diabetic cardiomy-
cial in maintaining glucose and FFA metabolism and cardiac
opathy in response to hyperglycemia and insulin resistance
function. Conversely, a deficiency in PPAR-γ signaling may
(Figure 2). Oxidative stress, inflammation, and enhanced
contribute to the development of diabetic cardiomyopathy.
RAAS and SNS activity further promote PKC activation. To
Increased O-GlcNAc in Promoting Cardiac Fibrosis date, ≈15 isoforms of PKC have been described in humans.
in Diabetic Cardiomyopathy These isoforms can be divided into 3 subfamilies based on
Hyperglycemia is associated with increased O-GlcNAcylation, second messenger signaling and particular mode of activa-
which causes post-translational modification of cardiac pro- tion.128,129 PKCα, β, ε, θ, and δ isoforms have been proposed
teins (Figure 2). Sustained O-GlcNAc signaling exists in the to be involved in the development of diabetic cardiac hyper-
diabetic heart and can exert detrimental effects that include trophy.128,129 For example, PKC β2 has been shown to medi-
decreases in mitochondrial function and energy generation ate hyperglycemia-induced diastolic cardiac dysfunction in
and increases in cardiac dysfunction and heart failure.116 diabetic rats through alterations in caveolin-3 expression and
Under physiological conditions, the hexosamine biosynthe- insulin metabolic Akt/eNOS signaling.128 Further supporting
sis pathway drives a component of fructose-6-phosphate me- its relevance, targeted inhibition of PKC β2 in a transgenic
tabolism from glycolysis to generate the O-GlcNAc moiety.117 mouse model of diabetic cardiomyopathy has been reported
Transient activation of O-GlcNAc signaling is normally cyto- to improve fractional shortening and reverse cardiac hyper-
protective and increases cell survival.118 In contrast to transient trophy and fibrosis.130 Collectively, the data suggest that ac-
upregulation, sustained elevation of O-GlcNAc signaling in tivation of PKC can induce cellular and functional changes
Jia et al   Diabetic Cardiomyopathy and Heart Failure   633

that contribute to the development of diabetic cardiomyopa- regulators undergo a variety of modifications that cause dis-
thy and heart failure. sociation of Nrf2 from Keap1. Free Nrf2 can then bind to
small Maf proteins in the nucleus to activate transcription.137
Role of MAPK and JNK Activation in the Genesis Hyperglycemia and insulin resistance repress Nrf2 expres-
of Diabetic Cardiomyopathy sion and activity through an Erk 1/2-mediated pathway that
MAPK activation has also been implicated in the pathogen- contributes to oxidative stress and insulin resistance in car-
esis of diabetic cardiomyopathy and heart failure. Erk1/2, diomyocytes.138 Restoration of Nrf2 activity prevents diabetes
p38 MAPK, and JNKs are 3 important MAPK subfamilies mellitus–induced lipid accumulation, inflammation, fibrosis,
that regulate cardiac growth, hypertrophy, and remodeling.13 and associated cardiac dysfunction,139 providing another po-
Enhanced myocardial phosphorylation of Erk 1/2 and activa- tential strategy for the prevention of diabetic cardiomyopathy.
tion of p38 MAPK occurs during ischemia in streptozotocin-
induced diabetic models.131 Our research and that of others Role of the Transcription Factor CREM in the
has demonstrated that obesity/insulin resistance–induced car- Genesis of Diabetic Cardiomyopathy
diac dysfunction is associated with enhanced S6 kinase 1 and CREM is a transcription factor that regulates cAMP signal-
Erk1/2 signaling.17,132 JNK can be activated by oxidative stress, ing and cardiac gene expression (Figure 2). Members of the
inflammatory cytokines, and sphingolipid metabolites.133 In CREM family may promote fibrosis of the heart, especially
turn, enhanced JNK signaling in the diabetic heart contributes in response to hyperglycemia and elevated FFAs.140 For ex-
to oxidative stress, endoplasmic reticulum stress, and intersti- ample, CREM expression is increased in cardiomyocytes of
tial fibrosis.133 In contrast, inhibition of JNK phosphorylation chronically hyperglycemic type 1 diabetic mice, and this is
by a curcumin analog prevents high glucose-induced inflam- accompanied by increased cardiac fibrosis.141 These adverse
mation and apoptosis in diabetic hearts.133 In addition to these effects were associated with alterations in histone acetylation
observations, JNK may play an important role in cardiomyo- and alterations in miRNAs profiles, suggesting that CREM
cyte apoptosis.134 As such, JNK activation was shown to cause activation may promote epigenetic and genetic modifications
increased cardiomyocyte apoptosis as early as days 3 and 7 in in cardiac proteins and mediate glycemic memory in ongoing
a type 1 diabetic rodent model.134 Collectively, activation of progression of diabetic cardiomyopathy.
both MAPK and JNK signaling seems to contribute signifi-
cantly to the development of diabetic cardiomyopathy. Role of miRNAs in Promotion of Diabetic
Cardiomyopathy
Role of NF-κB Activation in the Genesis of Diabetic Diabetic cardiomyopathy is associated with increased expres-
Cardiomyopathy sion of miRNAs—a group of short single-stranded noncod-
Downloaded from http://ahajournals.org by on February 10, 2022

NF-κB is one of the key transcription factors that regulate the ing RNA molecules with an average length of 22 nucleotides.
expression of proinflammatory cytokines, profibrotic genes, Importantly, miRNAs control the expression of transcriptional
and cell survival,135 thus contributing to mitochondrial and and post-transcriptional target genes through binding to the
cardiac dysfunction in diabetic hearts. NF-κB is found in the 3′-untranslated region and regulate mitochondrial function,
cytoplasm of nonstimulated cells. On stimulation, IκB is phos- ROS production, Ca2+ handling, apoptosis, autophagy, and fi-
phorylated and its p50/p65 subunits translocate to the nucleus brosis, all of which are regarded as important mechanisms in
and bind κB nuclear elements.135 In diabetes mellitus, ROS, diabetes mellitus–induced cardiac hypertrophy, remodeling,
AGEs, and an activated cardiac tissue RAAS can directly in- and fibrosis, as well as heart failure progression.142,143 miR-
duce NF-κB activation. This, in turn, promotes maladaptive 15a, -21, -24, -29, -30d, -103, -126, -146a, -150, -191, -223,
immune responses and the release of proinflammatory cyto- -320, -375, and -486 have all been reported to be increased in
kines, such as tumor necrosis factor α, monocyte chemotactic type 2 diabetic individuals.142,143 In cardiac tissue of a type 1
protein 1, interleukin-6, and interleukin-8.13 Activated NF-κB diabetic rodent model, expression of miR-21, -24, -142-3p,
in diabetic mouse hearts has been shown to be associated with -195, -199a-3p, -700, -705, -208, -221, and 499-3p was upreg-
increased nicotinamide adenine dinucleotide phosphate oxi- ulated whereas expression of miR-1, -20a, -29a, -143, -220b,
dase–mediated generation of ROS, peroxynitrite, and super- and -373 was downregulated.144 miR103, 107, -143, and -181
oxide.136 These processes lead to a reduction in bioavailable play a role in insulin sensitivity and systemic glucose metabo-
NO. Inhibition of NF-κB with pyrrolidine dithiocarbamate lism.145,146 Increased expression of miR-454, 500,-142-3p/5p,
improves mitochondrial structural integrity and inhibits and 1246 has been identified in cardiac diastolic dysfunc-
oxidative stress, increasing ATP synthesis and NO bioavail- tion.147 Other miRNAs, such as miR-113a, -133a, -150 have
ability, thereby restoring cardiac function in type 2 diabetes been found to be involved in the regulation of cardiomyocyte
mellitus.136 hypertrophy and interstitial fibrosis.148

Abnormalities of Nrf2-Related Antioxidant Actions Abnormalities of Exosomes in Diabetic


in Diabetic Cardiomyopathy Cardiomyopathy
Nrf2 is a leucine zipper protein that promotes the expression There is a close relationship between nutrient metabolism
of antioxidant proteins, such as hemoxygenase in response to and exosome release from cells, such as cardiomyocytes.
oxidative stress (Figure 2). Nrf2 is predominantly regulated Exosomes are extracellular vesicles with a diameter ranging
by its binding to the inhibitor, Keap1, which targets Nrf2 for from 30 to 90 nm and are regarded as important mediators
ubiquitination and degradation, thus maintaining low cel- of cell-to-cell communication.24 They contain a variety of
lular levels of Nrf2.137 Under oxidative stress, Nrf2 and its biological components, including lipids, miRNAs, proteins,
634  Circulation Research  February 16, 2018

and transcription factors that regulate both normal physiologi- does not improve cardiac event rates.152 Unfortunately, screen-
cal and pathophysiological effects.24 Exosomes that function ing approaches including B-type natriuretic peptide, exercise
with glucose transporters and glycolytic enzymes to increase stress testing, and echocardiographic assessment do not seem
glucose uptake, glycolysis, and pyruvate production in endo- to be sufficiently sensitive to identify subclinical dysfunction
thelial cells were released from cardiomyocytes after glucose in diabetic patients.153 Therefore, further studies are vital to
deprivation.149 In type 2 diabetic hearts, exosomes containing the understanding of the precise mechanisms involved in the
high levels of miR-320 are released from cardiomyocytes and initiation and progression of diabetic cardiomyopathy and to
transported to coronary endothelial cells, resulting in reduced the development of novel strategies to reduce the risk of heart
NO production and inhibition of angiogenesis via decreases failure in diabetic patients.
heat shock protein 20.150 However, heat shock protein 20–en-
gineered exosomes have a beneficial role in the regulation Sources of Funding
of cardiomyocyte exosome secretion and restoration of hy- J.R. Sowers received funding from National Institutes of Health
perglycemia-induced cardiac dysfunction.151 Therefore, exo- (NIH; R01 HL73101-01A and R01 HL107910-01) and the Veterans
Affairs Merit System (0018). Dr Jia received funding from American
somes may not only act as biomarkers, but targeting exosomes Diabetes Association (Innovative Basic Science Award no. 1-17-IBS-
may also be a potential therapeutic strategy in the prevention 201). Dr Hill received funding from NIH (RO1HL085119).
or progression of diabetic cardiomyopathy.
Disclosures
Conclusion and Future Perspectives None.
The cardinal features of diabetic cardiomyopathy include car-
diac stiffness, myocardial fibrosis, and hypertrophy with car- References
diac diastolic dysfunction and subsequent progression to both 1. Rubler S, Dlugash J, Yuceoglu YZ, Kumral T, Branwood AW, Grishman A.
New type of cardiomyopathy associated with diabetic glomerulosclerosis.
systolic dysfunction and clinical heart failure. Importantly, Am J Cardiol. 1972;30:595–602.
hyperglycemia and systemic and cardiac insulin resistance 2. Kannel WB, Hjortland M, Castelli WP. Role of diabetes in congestive
are independently associated with the development and pro- heart failure: the Framingham study. Am J Cardiol. 1974;34:29–34.
3. Yancy CW, Jessup M, Bozkurt B, et al; American College of Cardiology
gression of cardiac dysfunction and heart failure in diabetes
Foundation; American Heart Association Task Force on Practice
mellitus. From a mechanistic point of view, mitochondrial Guidelines. 2013 ACCF/AHA guideline for the management of heart fail-
dysfunction, oxidative stress, increased formation and depo- ure: a report of the American College of Cardiology Foundation/American
sition of AGEs, impaired mitochondrial Ca2+ handling and Heart Association Task Force on Practice Guidelines. J Am Coll Cardiol.
2013;62:e147–e239. doi: 10.1016/j.jacc.2013.05.019.
function, inflammation, activation of RAAS and SNS, cardiac
Downloaded from http://ahajournals.org by on February 10, 2022

4. Ryden L, Grant PJ, Anker SD, Berne C, Cosentino F, Danchin N, Deaton


autonomic neuropathy, endoplasmic reticulum stress, micro- C, Escaned J, Hammes HP, Huikuri H, Marre M, Marx N, Mellbin L,
vascular dysfunction, and cardiac metabolic disorders are in- Ostergren J, Patrono C, Seferovic P, Uva MS, Taskinen MR, Tendera
M, Tuomilehto J, Valensi P, Zamorano JL, Zamorano JL, Achenbach S,
volved in the pathophysiological process. Underlying these Baumgartner H, Bax JJ, Bueno H, Dean V, Deaton C, Erol C, Fagard R,
pathophysiological events, evidence supports a role for sever- Ferrari R, Hasdai D, Hoes AW, Kirchhof P, Knuuti J, Kolh P, Lancellotti
al proteins and signaling pathways, including AMPK, PPARs, P, Linhart A, Nihoyannopoulos P, Piepoli MF, Ponikowski P, Sirnes
O-GlcNAc, SGLT2, PKC, MAPK, NFκB, Nrf2, miRNA, and PA, Tamargo JL, Tendera M, Torbicki A, Wijns W, Windecker S, De
Backer G, Sirnes PA, Ezquerra EA, Avogaro A, Badimon L, Baranova
exosomes. Although diabetic cardiomyopathy seems to have E, Baumgartner H, Betteridge J, Ceriello A, Fagard R, Funck-Brentano
an extensive preclinical course and the pathophysiological C, Gulba DC, Hasdai D, Hoes AW, Kjekshus JK, Knuuti J, Kolh P, Lev
changes seem to be induced by the metabolic alterations in di- E, Mueller C, Neyses L, Nilsson PM, Perk J, Ponikowski P, Reiner Z,
Sattar N, Schachinger V, Scheen A, Schirmer H, Stromberg A, Sudzhaeva
abetes mellitus, a formal definition for diabetic cardiomyopa- S, Tamargo JL, Viigimaa M, Vlachopoulos C, Xuereb RG; Authors/
thy as a distinct clinical entity remains vague because of a lack Task Force Members; ESC Committee for Practice Guidelines (CPG);
of accepted diagnostic criteria and information on subclinical Document Reviewers. ESC Guidelines on diabetes, pre-diabetes, and
CVD in the early stages of diabetes mellitus. Currently, there cardiovascular diseases developed in collaboration with the EASD: the
Task Force on diabetes, pre-diabetes, and cardiovascular diseases of the
is not a specific histological property, biochemical marker, or European Society of Cardiology (ESC) and developed in collaboration
clinical manifestation for the definitive diagnosis of diabetic with the European Association for the Study of Diabetes (EASD). Eur
cardiomyopathy. Also, there are no prospective clinical trials Heart J. 2013;34:3035–3087. doi: 10.1093/eurheartj/eht108.
5. Rydén L, Armstrong PW, Cleland JG, Horowitz JD, Massie BM, Packer
to support that hyperglycemia or hyperinsulinemia indepen- M, Poole-Wilson PA. Efficacy and safety of high-dose lisinopril in chronic
dently increase the risk for development of diabetic cardio- heart failure patients at high cardiovascular risk, including those with dia-
myopathy in the absence of other risk factors, such as obesity, betes mellitus. Results from the ATLAS trial. Eur Heart J. 2000;21:1967–
coronary heart disease, and hypertension. Recent scientific ev- 1978. doi: 10.1053/euhj.2000.2311.
6. Shindler DM, Kostis JB, Yusuf S, Quinones MA, Pitt B, Stewart D, Pinkett
idence for the potential use of exosome and circulating miR- T, Ghali JK, Wilson AC. Diabetes mellitus, a predictor of morbidity and
NAs as biomarkers for detection of diabetic cardiomyopathy mortality in the Studies of Left Ventricular Dysfunction (SOLVD) Trials
highlights emerging methods for the diagnosis and preven- and Registry. Am J Cardiol. 1996;77:1017–1020.
7. Thrainsdottir IS, Aspelund T, Thorgeirsson G, Gudnason V, Hardarson T,
tion of diabetes mellitus and cardiomyopathy. The American Malmberg K, Sigurdsson G, Rydén L. The association between glucose
Diabetes Association recommends screening patients with abnormalities and heart failure in the population-based Reykjavik study.
type 2 diabetes mellitus for the prevention of diabetic cardio- Diabetes Care. 2005;28:612–616.
8. Aronow WS, Ahn C. Incidence of heart failure in 2,737 older persons with
myopathy, but the results of the recently published Detection
and without diabetes mellitus. Chest. 1999;115:867–868.
of Ischemia in Asymptomatic Diabetics trial indicate that 9. Devereux RB, Roman MJ, Paranicas M, O’Grady MJ, Lee ET, Welty TK,
screening of asymptomatic patients with nuclear imaging Fabsitz RR, Robbins D, Rhoades ER, Howard BV. Impact of diabetes
Jia et al   Diabetic Cardiomyopathy and Heart Failure   635

on cardiac structure and function: the strong heart study. Circulation. heterozygous SERCA2a knockout mice. Am J Physiol Heart Circ Physiol.
2000;101:2271–2276. 2008;294:H1426–H1434. doi: 10.1152/ajpheart.01016.2007.
10. Bertoni AG, Goff DC Jr, D’Agostino RB Jr, Liu K, Hundley WG, Lima 26. Pollack PS, Malhotra A, Fein FS, Scheuer J. Effects of diabetes on car-
JA, Polak JF, Saad MF, Szklo M, Tracy RP, Siscovick DS. Diabetic cardio- diac contractile proteins in rabbits and reversal with insulin. Am J Physiol.
myopathy and subclinical cardiovascular disease: the Multi-Ethnic Study 1986;251:H448–H454. doi: 10.1152/ajpheart.1986.251.2.H448.
of Atherosclerosis (MESA). Diabetes Care. 2006;29:588–594. 27. Malhotra A, Sanghi V. Regulation of contractile proteins in diabetic heart.
11. Lind M, Bounias I, Olsson M, Gudbjörnsdottir S, Svensson AM, Rosengren Cardiovasc Res. 1997;34:34–40.
A. Glycaemic control and incidence of heart failure in 20,985 patients with 28. Kanamori H, Takemura G, Goto K, Tsujimoto A, Mikami A, Ogino

type 1 diabetes: an observational study. Lancet. 2011;378:140–146. doi: A, Watanabe T, Morishita K, Okada H, Kawasaki M, Seishima M,
10.1016/S0140-6736(11)60471-6. Minatoguchi S. Autophagic adaptations in diabetic cardiomyopathy differ
12. Stratton IM, Adler AI, Neil HA, Matthews DR, Manley SE, Cull CA, between type 1 and type 2 diabetes. Autophagy. 2015;11:1146–1160. doi:
Hadden D, Turner RC, Holman RR. Association of glycaemia with mac- 10.1080/15548627.2015.1051295.
rovascular and microvascular complications of type 2 diabetes (UKPDS 29. Holscher ME, Bode C, Bugger H. Diabetic cardiomyopathy: does the
35): prospective observational study. BMJ. 2000;321:405–412. type of diabetes matter? Int J Mol Sci. 2016;17:E2136. doi: 10.3390/
13. Jia G, DeMarco VG, Sowers JR. Insulin resistance and hyperinsulinaemia ijms17122136.
in diabetic cardiomyopathy. Nat Rev Endocrinol. 2016;12:144–153. doi: 30. Bugger H, Boudina S, Hu XX, Tuinei J, Zaha VG, Theobald HA, Yun UJ,
10.1038/nrendo.2015.216. McQueen AP, Wayment B, Litwin SE, Abel ED. Type 1 diabetic Akita
14. Itzhaki Ben Zadok O, Kornowski R, Goldenberg I, Klempfner R, Toledano mouse hearts are insulin sensitive but manifest structurally abnormal mi-
Y, Biton Y, Fisman EZ, Tenenbaum A, Golovchiner G, Kadmon E, tochondria that remain coupled despite increased uncoupling protein 3.
Omelchenko A, Gal TB, Barsheshet A. Admission blood glucose and 10- Diabetes. 2008;57:2924–2932.
year mortality among patients with or without pre-existing diabetes mel- 31. Rosenkranz AC, Hood SG, Woods RL, Dusting GJ, Ritchie RH. B-type
litus hospitalized with heart failure. Cardiovasc Diabetol. 2017;16:102. natriuretic peptide prevents acute hypertrophic responses in the diabetic
doi: 10.1186/s12933-017-0582-y. rat heart: importance of cyclic GMP. Diabetes. 2003;52:2389–2395.
15. Bahrami H, Bluemke DA, Kronmal R, Bertoni AG, Lloyd-Jones DM, 32. Sundgren NC, Giraud GD, Schultz JM, Lasarev MR, Stork PJ, Thornburg
Shahar E, Szklo M, Lima JA. Novel metabolic risk factors for incident KL. Extracellular signal-regulated kinase and phosphoinositol-3 kinase
heart failure and their relationship with obesity: the MESA (Multi-Ethnic mediate IGF-1 induced proliferation of fetal sheep cardiomyocytes. Am
Study of Atherosclerosis) study. J Am Coll Cardiol. 2008;51:1775–1783. J Physiol Regul Integr Comp Physiol. 2003;285:R1481–R1489. doi:
doi: 10.1016/j.jacc.2007.12.048. 10.1152/ajpregu.00232.2003.
16. Mellor KM, Bell JR, Young MJ, Ritchie RH, Delbridge LM. Myocardial 33. Wang J, Song Y, Wang Q, Kralik PM, Epstein PN. Causes and character-
autophagy activation and suppressed survival signaling is associated with istics of diabetic cardiomyopathy. Rev Diabet Stud. 2006;3:108–117. doi:
insulin resistance in fructose-fed mice. J Mol Cell Cardiol. 2011;50:1035– 10.1900/RDS.2006.3.108.
1043. doi: 10.1016/j.yjmcc.2011.03.002. 34. Mytas DZ, Stougiannos PN, Zairis MN, Foussas SG, Pyrgakis VN,

17. Jia G, Habibi J, DeMarco VG, Martinez-Lemus LA, Ma L, Whaley- Kyriazis IA. Diabetic myocardial disease: pathophysiology, early diagno-
Connell AT, Aroor AR, Domeier TL, Zhu Y, Meininger GA, Mueller KB, sis and therapeutic options. J Diabetes Complications. 2009;23:273–282.
Jaffe IZ, Sowers JR. Endothelial mineralocorticoid receptor deletion pre- doi: 10.1016/j.jdiacomp.2007.12.005.
vents diet-induced cardiac diastolic dysfunction in females. Hypertension. 35. Bando YK, Murohara T. Diabetes-related heart failure. Circ J.

2015;66:1159–1167. doi: 10.1161/HYPERTENSIONAHA.115.06015. 2014;78:576–583.
Downloaded from http://ahajournals.org by on February 10, 2022

18. Lee M, Gardin JM, Lynch JC, Smith VE, Tracy RP, Savage PJ, Szklo M, 36. Passino C, Barison A, Vergaro G, Gabutti A, Borrelli C, Emdin M, Clerico
Ward BJ. Diabetes mellitus and echocardiographic left ventricular func- A. Markers of fibrosis, inflammation, and remodeling pathways in heart
tion in free-living elderly men and women: the Cardiovascular Health failure. Clin Chim Acta. 2015;443:29–38. doi: 10.1016/j.cca.2014.09.006.
Study. Am Heart J. 1997;133:36–43. 37. Bugger H, Riehle C, Jaishy B, Wende AR, Tuinei J, Chen D, Soto J, Pires
19. Tate M, Deo M, Cao AH, Hood SG, Huynh K, Kiriazis H, Du XJ, Julius KM, Boudina S, Theobald HA, Luptak I, Wayment B, Wang X, Litwin SE,
TL, Figtree GA, Dusting GJ, Kaye DM, Ritchie RH. Insulin replacement Weimer BC, Abel ED. Genetic loss of insulin receptors worsens cardiac
limits progression of diabetic cardiomyopathy in the low-dose strepto- efficiency in diabetes. J Mol Cell Cardiol. 2012;52:1019–1026.
zotocin-induced diabetic rat. Diab Vasc Dis Res. 2017;14:423–433. doi: 38. Qi Y, Xu Z, Zhu Q, Thomas C, Kumar R, Feng H, Dostal DE, White MF,
10.1177/1479164117710390. Baker KM, Guo S. Myocardial loss of IRS1 and IRS2 causes heart fail-
20. Andersson C, Olesen JB, Hansen PR, Weeke P, Norgaard ML, Jørgensen ure and is controlled by p38α MAPK during insulin resistance. Diabetes.
CH, Lange T, Abildstrøm SZ, Schramm TK, Vaag A, Køber L, Torp- 2013;62:3887–3900. doi: 10.2337/db13-0095.
Pedersen C, Gislason GH. Metformin treatment is associated with a low 39. Cook SA, Varela-Carver A, Mongillo M, Kleinert C, Khan MT, Leccisotti
risk of mortality in diabetic patients with heart failure: a retrospective na- L, Strickland N, Matsui T, Das S, Rosenzweig A, Punjabi P, Camici
tionwide cohort study. Diabetologia. 2010;53:2546–2553. doi: 10.1007/ PG. Abnormal myocardial insulin signalling in type 2 diabetes and left-
s00125-010-1906-6. ventricular dysfunction. Eur Heart J. 2010;31:100–111. doi: 10.1093/
21. Zinman B, Wanner C, Lachin JM, Fitchett D, Bluhmki E, Hantel S, eurheartj/ehp396.
Mattheus M, Devins T, Johansen OE, Woerle HJ, Broedl UC, Inzucchi 40. Song R, Peng W, Zhang Y, et al. Central role of E3 ubiquitin ligase MG53
SE; EMPA-REG OUTCOME Investigators. Empagliflozin, cardio- in insulin resistance and metabolic disorders. Nature. 2013;494:375–379.
vascular outcomes, and mortality in type 2 diabetes. N Engl J Med. doi: 10.1038/nature11834.
2015;373:2117–2128. doi: 10.1056/NEJMoa1504720. 41. Liu F, Song R, Feng Y, Guo J, Chen Y, Zhang Y, Chen T, Wang Y, Huang
22. Marso SP, Bain SC, Consoli A, Eliaschewitz FG, Jódar E, Leiter LA, Y, Li CY, Cao C, Zhang Y, Hu X, Xiao RP. Upregulation of MG53 induces
Lingvay I, Rosenstock J, Seufert J, Warren ML, Woo V, Hansen O, Holst diabetic cardiomyopathy through transcriptional activation of peroxisome
AG, Pettersson J, Vilsbøll T; SUSTAIN-6 Investigators. Semaglutide and proliferation-activated receptor α. Circulation. 2015;131:795–804. doi:
cardiovascular outcomes in patients with type 2 diabetes. N Engl J Med. 10.1161/CIRCULATIONAHA.114.012285.
2016;375:1834–1844. doi: 10.1056/NEJMoa1607141. 42. Vincent MA, Clerk LH, Lindner JR, Klibanov AL, Clark MG, Rattigan S,
23. Marso SP, Daniels GH, Brown-Frandsen K, Kristensen P, Mann JF, Nauck Barrett EJ. Microvascular recruitment is an early insulin effect that regu-
MA, Nissen SE, Pocock S, Poulter NR, Ravn LS, Steinberg WM, Stockner lates skeletal muscle glucose uptake in vivo. Diabetes. 2004;53:1418–1423.
M, Zinman B, Bergenstal RM, Buse JB; LEADER Steering Committee; 43. Vincent MA, Clerk LH, Lindner JR, Price WJ, Jahn LA, Leong-Poi H,
LEADER Trial Investigators. Liraglutide and cardiovascular outcomes Barrett EJ. Mixed meal and light exercise each recruit muscle capillaries
in type 2 diabetes. N Engl J Med. 2016;375:311–322. doi: 10.1056/ in healthy humans. Am J Physiol Endocrinol Metab. 2006;290:E1191–
NEJMoa1603827. E1197. doi: 10.1152/ajpendo.00497.2005.
24. Westermeier F, Riquelme JA, Pavez M, Garrido V, Díaz A, Verdejo HE, 44. Vollus GC, Bradley EA, Roberts MK, Newman JM, Richards SM,

Castro PF, García L, Lavandero S. New molecular insights of insulin Rattigan S, Barrett EJ, Clark MG. Graded occlusion of perfused rat muscle
in diabetic cardiomyopathy. Front Physiol. 2016;7:125. doi: 10.3389/ vasculature decreases insulin action. Clin Sci (Lond). 2007;112:457–466.
fphys.2016.00125. doi: 10.1042/CS20060311.
25. Talukder MA, Kalyanasundaram A, Zuo L, Velayutham M, Nishijima Y, 45. Jones SP, Bolli R. The ubiquitous role of nitric oxide in cardioprotec-
Periasamy M, Zweier JL. Is reduced SERCA2a expression detrimental tion. J Mol Cell Cardiol. 2006;40:16–23. doi: 10.1016/j.yjmcc.2005.
or beneficial to postischemic cardiac function and injury? Evidence from 09.011.
636  Circulation Research  February 16, 2018

46. Battiprolu PK, Hojayev B, Jiang N, Wang ZV, Luo X, Iglewski M,


A unifying hypothesis. Diabetes Care. 2016;39:1115–1122. doi: 10.2337/
Shelton JM, Gerard RD, Rothermel BA, Gillette TG, Lavandero S, Hill dc16-0542.
JA. Metabolic stress-induced activation of FoxO1 triggers diabetic car- 67. Hue L, Taegtmeyer H. The Randle cycle revisited: a new head for an old
diomyopathy in mice. J Clin Invest. 2012;122:1109–1118. doi: 10.1172/ hat. Am J Physiol Endocrinol Metab. 2009;297:E578–E591. doi: 10.1152/
JCI60329. ajpendo.00093.2009.
47. Qi Y, Zhu Q, Zhang K, Thomas C, Wu Y, Kumar R, Baker KM, Xu Z, 68. Bowker-Kinley MM, Davis WI, Wu P, Harris RA, Popov KM. Evidence
Chen S, Guo S. Activation of Foxo1 by insulin resistance promotes cardiac for existence of tissue-specific regulation of the mammalian pyruvate de-
dysfunction and β-myosin heavy chain gene expression. Circ Heart Fail. hydrogenase complex. Biochem J. 1998;329(pt 1):191–196.
2015;8:198–208. doi: 10.1161/CIRCHEARTFAILURE.114.001457. 69. Randle PJ, Garland PB, Hales CN, Newsholme EA. The glucose fatty-
48. Mellor KM, Bell JR, Ritchie RH, Delbridge LM. Myocardial insulin re- acid cycle. Its role in insulin sensitivity and the metabolic disturbances of
sistance, metabolic stress and autophagy in diabetes. Clin Exp Pharmacol diabetes mellitus. Lancet. 1963;1:785–789.
Physiol. 2013;40:56–61. doi: 10.1111/j.1440-1681.2012.05738.x. 70. McGarry JD. What if Minkowski had been ageusic? An alternative angle
49. Samuel VT. Fructose induced lipogenesis: from sugar to fat to insulin on diabetes. Science. 1992;258:766–770.
resistance. Trends Endocrinol Metab. 2011;22:60–65. doi: 10.1016/j. 71. Kim JA, Wei Y, Sowers JR. Role of mitochondrial dysfunction in insulin
tem.2010.10.003. resistance. Circ Res. 2008;102:401–414. doi: 10.1161/CIRCRESAHA.
50. Johnson RJ, Sanchez-Lozada LG, Nakagawa T. The effect of fructose on 107.165472.
renal biology and disease. J Am Soc Nephrol. 2010;21:2036–2039. doi: 72. Anderson EJ, Kypson AP, Rodriguez E, Anderson CA, Lehr EJ, Neufer
10.1681/ASN.2010050506. PD. Substrate-specific derangements in mitochondrial metabolism and re-
51. Delbridge LM, Benson VL, Ritchie RH, Mellor KM. Diabetic cardio- dox balance in the atrium of the type 2 diabetic human heart. J Am Coll
myopathy: the case for a role of fructose in disease etiology. Diabetes. Cardiol. 2009;54:1891–1898. doi: 10.1016/j.jacc.2009.07.031.
2016;65:3521–3528. doi: 10.2337/db16-0682. 73. Anderson EJ, Rodriguez E, Anderson CA, Thayne K, Chitwood

52. Vega RB, Horton JL, Kelly DP. Maintaining ancient organelles: mito- WR, Kypson AP. Increased propensity for cell death in diabetic hu-
chondrial biogenesis and maturation. Circ Res. 2015;116:1820–1834. doi: man heart is mediated by mitochondrial-dependent pathways. Am
10.1161/CIRCRESAHA.116.305420. J Physiol Heart Circ Physiol. 2011;300:H118–H124. doi: 10.1152/
53. Guo CA, Guo S. Insulin receptor substrate signaling controls cardiac en- ajpheart.00932.2010.
ergy metabolism and heart failure. J Endocrinol. 2017;233:R131–R143. 74. Teshima Y, Takahashi N, Nishio S, Saito S, Kondo H, Fukui A, Aoki K,
doi: 10.1530/JOE-16-0679. Yufu K, Nakagawa M, Saikawa T. Production of reactive oxygen species
54. Lee TW, Bai KJ, Lee TI, Chao TF, Kao YH, Chen YJ. PPARs modulate in the diabetic heart. Roles of mitochondria and NADPH oxidase. Circ J.
cardiac metabolism and mitochondrial function in diabetes. J Biomed Sci. 2014;78:300–306.
2017;24:5. doi: 10.1186/s12929-016-0309-5. 75. Jia G, Habibi J, Aroor AR, Hill MA, DeMarco VG, Lee LE, Ma L,
55. Habets DD, Coumans WA, Voshol PJ, den Boer MA, Febbraio M,
Barron BJ, Whaley-Connell A, Sowers JR. Enhanced endothelium epithe-
Bonen A, Glatz JF, Luiken JJ. AMPK-mediated increase in myocardial lial sodium channel signaling prompts left ventricular diastolic dysfunc-
long-chain fatty acid uptake critically depends on sarcolemmal CD36. tion in obese female mice. Metabolism. 2018;78:69–79. doi: 10.1016/j.
Biochem Biophys Res Commun. 2007;355:204–210. doi: 10.1016/j. metabol.2017.08.008.
bbrc.2007.01.141. 76. Murdoch CE, Chaubey S, Zeng L, Yu B, Ivetic A, Walker SJ, Vanhoutte
56. Yang J, Sambandam N, Han X, Gross RW, Courtois M, Kovacs A,
D, Heymans S, Grieve DJ, Cave AC, Brewer AC, Zhang M, Shah AM.
Febbraio M, Finck BN, Kelly DP. CD36 deficiency rescues lipotoxic Endothelial NADPH oxidase-2 promotes interstitial cardiac fibrosis and
Downloaded from http://ahajournals.org by on February 10, 2022

cardiomyopathy. Circ Res. 2007;100:1208–1217. doi: 10.1161/01. diastolic dysfunction through proinflammatory effects and endothelial-
RES.0000264104.25265.b6. mesenchymal transition. J Am Coll Cardiol. 2014;63:2734–2741. doi:
57. Tanaka T, Nakata T, Oka T, Ogawa T, Okamoto F, Kusaka Y, Sohmiya 10.1016/j.jacc.2014.02.572.
K, Shimamoto K, Itakura K. Defect in human myocardial long-chain 77. Lee SJ, Kang JG, Ryu OH, Kim CS, Ihm SH, Choi MG, Yoo HJ, Kim
fatty acid uptake is caused by FAT/CD36 mutations. J Lipid Res. DS, Kim TW. Effects of alpha-lipoic acid on transforming growth fac-
2001;42:751–759. tor beta1-p38 mitogen-activated protein kinase-fibronectin pathway in
58. Zou MH, Xie Z. Regulation of interplay between autophagy and apoptosis diabetic nephropathy. Metabolism. 2009;58:616–623. doi: 10.1016/j.
in the diabetic heart: new role of AMPK. Autophagy. 2013;9:624–625. metabol.2008.12.006.
doi: 10.4161/auto.23577. 78. Giacco F, Brownlee M. Oxidative stress and diabetic complications. Circ
59. Finck BN, Lehman JJ, Leone TC, Welch MJ, Bennett MJ, Kovacs A, Han Res. 2010;107:1058–1070. doi: 10.1161/CIRCRESAHA.110.223545.
X, Gross RW, Kozak R, Lopaschuk GD, Kelly DP. The cardiac phenotype 79. Raposeiras-Roubín S, Rodiño-Janeiro BK, Paradela-Dobarro B, Grigorian-
induced by PPARalpha overexpression mimics that caused by diabetes Shamagian L, García-Acuña JM, Aguiar-Souto P, Jacquet-Hervet M,
mellitus. J Clin Invest. 2002;109:121–130. doi: 10.1172/JCI14080. Reino-Maceiras MV, Alvarez E, González-Juanatey JR. Predictive value
60. Samuel VT, Shulman GI. The pathogenesis of insulin resistance: integrat- of advanced glycation end products for the development of post-infarction
ing signaling pathways and substrate flux. J Clin Invest. 2016;126:12–22. heart failure: a preliminary report. Cardiovasc Diabetol. 2012;11:102. doi:
doi: 10.1172/JCI77812. 10.1186/1475-2840-11-102.
61. Jornayvaz FR, Birkenfeld AL, Jurczak MJ, Kanda S, Guigni BA, Jiang 80. Willemsen S, Hartog JW, van Veldhuisen DJ, van der Meer P, Roze JF,
DC, Zhang D, Lee HY, Samuel VT, Shulman GI. Hepatic insulin resis- Jaarsma T, Schalkwijk C, van der Horst IC, Hillege HL, Voors AA. The
tance in mice with hepatic overexpression of diacylglycerol acyltrans- role of advanced glycation end-products and their receptor on outcome
ferase 2. Proc Natl Acad Sci USA. 2011;108:5748–5752. doi: 10.1073/ in heart failure patients with preserved and reduced ejection fraction. Am
pnas.1103451108. Heart J. 2012;164:742.e3–749.e3. doi: 10.1016/j.ahj.2012.07.027.
62. Atkinson LL, Kozak R, Kelly SE, Onay Besikci A, Russell JC, Lopaschuk 81. Liu W, Chen P, Deng J, Lv J, Liu J. Resveratrol and polydatin as modula-
GD. Potential mechanisms and consequences of cardiac triacylglycerol tors of Ca2+ mobilization in the cardiovascular system. Ann N Y Acad Sci.
accumulation in insulin-resistant rats. Am J Physiol Endocrinol Metab. 2017;1403:82–91. doi: 10.1111/nyas.13386.
2003;284:E923–E930. doi: 10.1152/ajpendo.00360.2002. 82. Kanaporis G, Blatter LA. Membrane potential determines calcium alter-
63. Cantley JL, Yoshimura T, Camporez JP, et al. CGI-58 knockdown se- nans through modulation of SR Ca2+ load and L-type Ca2+ current. J Mol
questers diacylglycerols in lipid droplets/ER-preventing diacylglyc- Cell Cardiol. 2017;105:49–58. doi: 10.1016/j.yjmcc.2017.02.004.
erol-mediated hepatic insulin resistance. Proc Natl Acad Sci USA. 83. Van den Bergh A, Vanderper A, Vangheluwe P, Desjardins F, Nevelsteen
2013;110:1869–1874. doi: 10.1073/pnas.1219456110. I, Verreth W, Wuytack F, Holvoet P, Flameng W, Balligand JL, Herijgers
64. Kruljac I, Ćaćić M, Ćaćić P, Ostojić V, Štefanović M, Šikić A, Vrkljan M. P. Dyslipidaemia in type II diabetic mice does not aggravate contrac-
Diabetic ketosis during hyperglycemic crisis is associated with decreased tile impairment but increases ventricular stiffness. Cardiovasc Res.
all-cause mortality in patients with type 2 diabetes mellitus. Endocrine. 2008;77:371–379. doi: 10.1093/cvr/cvm001.
2017;55:139–143. doi: 10.1007/s12020-016-1082-7. 84. Belke DD, Swanson EA, Dillmann WH. Decreased sarcoplasmic reticu-
65. Pawlak M, Baugé E, Lalloyer F, Lefebvre P, Staels B. Ketone body therapy lum activity and contractility in diabetic db/db mouse heart. Diabetes.
protects from lipotoxicity and acute liver failure upon PPARα deficiency. 2004;53:3201–3208.
Mol Endocrinol. 2015;29:1134–1143. doi: 10.1210/me.2014-1383. 85. Ye G, Metreveli NS, Ren J, Epstein PN. Metallothionein prevents diabe-
66. Mudaliar S, Alloju S, Henry RR. Can a shift in fuel energetics explain the tes-induced deficits in cardiomyocytes by inhibiting reactive oxygen spe-
beneficial cardiorenal outcomes in the EMPA-REG OUTCOME Study? cies production. Diabetes. 2003;52:777–783.
Jia et al   Diabetic Cardiomyopathy and Heart Failure   637

86. Ye G, Metreveli NS, Donthi RV, Xia S, Xu M, Carlson EC, Epstein PN. 105. Murthy VL, Naya M, Foster CR, Gaber M, Hainer J, Klein J, Dorbala
Catalase protects cardiomyocyte function in models of type 1 and type 2 S, Blankstein R, Di Carli MF. Association between coronary vascu-
diabetes. Diabetes. 2004;53:1336–1343. lar dysfunction and cardiac mortality in patients with and without
87. Uchimura K, Hayata M, Mizumoto T, et al. The serine protease prostasin diabetes mellitus. Circulation. 2012;126:1858–1868. doi: 10.1161/
regulates hepatic insulin sensitivity by modulating TLR4 signalling. Nat CIRCULATIONAHA.112.120402.
Commun. 2014;5:3428. doi: 10.1038/ncomms4428. 106. Garg R, Rao AD, Baimas-George M, Hurwitz S, Foster C, Shah RV,
88. Pal PB, Sonowal H, Shukla K, Srivastava SK, Ramana KV. Aldose re- Jerosch-Herold M, Kwong RY, Di Carli MF, Adler GK. Mineralocorticoid
ductase mediates NLRP3 inflammasome-initiated innate immune re- receptor blockade improves coronary microvascular function in indi-
sponse in hyperglycemia-induced Thp1 monocytes and male mice. viduals with type 2 diabetes. Diabetes. 2015;64:236–242. doi: 10.2337/
Endocrinology. 2017;158:3661–3675. doi: 10.1210/en.2017-00294. db14-0670.
89. Xu YZ, Zhang X, Wang L, Zhang F, Qiu Q, Liu ML, Zhang GR, Wu 107. Shome JS, Perera D, Plein S, Chiribiri A. Current perspectives in coro-
XL. An increased circulating angiotensin II concentration is associated nary microvascular dysfunction. Microcirculation. 2017;24:e12340. doi:
with hypoadiponectinemia and postprandial hyperglycemia in men with 10.1111/micc.12340.
nonalcoholic fatty liver disease. Intern Med. 2013;52:855–861. 108. Widyantoro B, Emoto N, Nakayama K, Anggrahini DW, Adiarto S, Iwasa
90. Giacchetti G, Sechi LA, Rilli S, Carey RM. The renin-angiotensin-al- N, Yagi K, Miyagawa K, Rikitake Y, Suzuki T, Kisanuki YY, Yanagisawa
dosterone system, glucose metabolism and diabetes. Trends Endocrinol M, Hirata K. Endothelial cell-derived endothelin-1 promotes cardiac
Metab. 2005;16:120–126. doi: 10.1016/j.tem.2005.02.003. fibrosis in diabetic hearts through stimulation of endothelial-to-mes-
91. Frantz EDC, Giori IG, Machado MV, Magliano DC, Freitas FM, Andrade enchymal transition. Circulation. 2010;121:2407–2418. doi: 10.1161/
MSB, Vieira AB, Nóbrega ACL, Tibiriçá E. High, but not low, exercise CIRCULATIONAHA.110.938217.
volume shifts the balance of renin-angiotensin system toward ACE2/Mas 109. Abdel Malik R, Zippel N, Frömel T, Heidler J, Zukunft S, Walzog B,
receptor axis in skeletal muscle in obese rats. Am J Physiol Endocrinol Ansari N, Pampaloni F, Wingert S, Rieger MA, Wittig I, Fisslthaler B,
Metab. 2017;313:E473–E482. doi: 10.1152/ajpendo.00078.2017. Fleming I. AMP-activated protein kinase α2 in neutrophils regulates vas-
92. Muñoz MC, Burghi V, Miquet JG, Cervino IA, Quiroga DT, Mazziotta L, cular repair via hypoxia-inducible factor-1α and a network of proteins
Dominici FP. Chronic blockade of the AT2 receptor with PD123319 im- affecting metabolism and apoptosis. Circ Res. 2017;120:99–109. doi:
pairs insulin signaling in C57BL/6 mice. Peptides. 2017;88:37–45. doi: 10.1161/CIRCRESAHA.116.309937.
10.1016/j.peptides.2016.12.003. 110. Lee TI, Kao YH, Chen YC, Huang JH, Hsiao FC, Chen YJ. Peroxisome
93. Miller JA, Floras JS, Zinman B, Skorecki KL, Logan AG. Effect of hy- proliferator-activated receptors modulate cardiac dysfunction in dia-
perglycaemia on arterial pressure, plasma renin activity and renal func- betic cardiomyopathy. Diabetes Res Clin Pract. 2013;100:330–339. doi:
tion in early diabetes. Clin Sci (Lond). 1996;90:189–195. 10.1016/j.diabres.2013.01.008.
94. Miller JA. Impact of hyperglycemia on the renin angiotensin sys- 111. Leone TC, Weinheimer CJ, Kelly DP. A critical role for the peroxisome
tem in early human type 1 diabetes mellitus. J Am Soc Nephrol. proliferator-activated receptor alpha (PPARalpha) in the cellular fasting
1999;10:1778–1785. response: the PPARalpha-null mouse as a model of fatty acid oxidation
95. Baudrand R, Gupta N, Garza AE, Vaidya A, Leopold JA, Hopkins PN, disorders. Proc Natl Acad Sci USA. 1999;96:7473–7478.
Jeunemaitre X, Ferri C, Romero JR, Williams J, Loscalzo J, Adler 112. Young ME, Patil S, Ying J, Depre C, Ahuja HS, Shipley GL, Stepkowski
GK, Williams GH, Pojoga LH. Caveolin 1 modulates aldosterone-me- SM, Davies PJ, Taegtmeyer H. Uncoupling protein 3 transcription is reg-
diated pathways of glucose and lipid homeostasis. J Am Heart Assoc. ulated by peroxisome proliferator-activated receptor (alpha) in the adult
2016;5:e003845. doi: 10.1161/JAHA.116.003845. rodent heart. FASEB J. 2001;15:833–845. doi: 10.1096/fj.00-0351com.
Downloaded from http://ahajournals.org by on February 10, 2022

96. Kim JA, Jang HJ, Martinez-Lemus LA, Sowers JR. Activation of mTOR/ 113. Zhou YT, Grayburn P, Karim A, Shimabukuro M, Higa M, Baetens D,
p70S6 kinase by ANG II inhibits insulin-stimulated endothelial ni- Orci L, Unger RH. Lipotoxic heart disease in obese rats: implications for
tric oxide synthase and vasodilation. Am J Physiol Endocrinol Metab. human obesity. Proc Natl Acad Sci USA. 2000;97:1784–1789.
2012;302:E201–E208. doi: 10.1152/ajpendo.00497.2011. 114. Razeghi P, Young ME, Cockrill TC, Frazier OH, Taegtmeyer H.

97. Pappachan JM, Sebastian J, Bino BC, Jayaprakash K, Vijayakumar K, Downregulation of myocardial myocyte enhancer factor 2C and myocyte
Sujathan P, Adinegara LA. Cardiac autonomic neuropathy in diabetes enhancer factor 2C-regulated gene expression in diabetic patients with
mellitus: prevalence, risk factors and utility of corrected QT interval nonischemic heart failure. Circulation. 2002;106:407–411.
in the ECG for its diagnosis. Postgrad Med J. 2008;84:205–210. doi: 115. Cheng L, Ding G, Qin Q, Huang Y, Lewis W, He N, Evans RM,

10.1136/pgmj.2007.064048. Schneider MD, Brako FA, Xiao Y, Chen YE, Yang Q. Cardiomyocyte-
98. Bisognano JD, Weinberger HD, Bohlmeyer TJ, Pende A, Raynolds MV, restricted peroxisome proliferator-activated receptor-delta deletion per-
Sastravaha A, Roden R, Asano K, Blaxall BC, Wu SC, Communal C, turbs myocardial fatty acid oxidation and leads to cardiomyopathy. Nat
Singh K, Colucci W, Bristow MR, Port DJ. Myocardial-directed over- Med. 2004;10:1245–1250. doi: 10.1038/nm1116.
expression of the human beta(1)-adrenergic receptor in transgenic mice. 116. Dassanayaka S, Jones SP. O-GlcNAc and the cardiovascular sys-

J Mol Cell Cardiol. 2000;32:817–830. doi: 10.1006/jmcc.2000.1123. tem. Pharmacol Ther. 2014;142:62–71. doi: 10.1016/j.pharmthera.
99. Axelrod S, Lishner M, Oz O, Bernheim J, Ravid M. Spectral analysis of 2013.11.005.
fluctuations in heart rate: an objective evaluation of autonomic nervous 117. Huynh K, Bernardo BC, McMullen JR, Ritchie RH. Diabetic cardiomy-
control in chronic renal failure. Nephron. 1987;45:202–206. opathy: mechanisms and new treatment strategies targeting antioxidant
100. Olshansky B, Sabbah HN, Hauptman PJ, Colucci WS. Parasympathetic signaling pathways. Pharmacol Ther. 2014;142:375–415. doi: 10.1016/j.
nervous system and heart failure: pathophysiology and potential im- pharmthera.2014.01.003.
plications for therapy. Circulation. 2008;118:863–871. doi: 10.1161/ 118. Hart GW, Housley MP, Slawson C. Cycling of O-linked beta-N-acetyl-
CIRCULATIONAHA.107.760405. glucosamine on nucleocytoplasmic proteins. Nature. 2007;446:1017–
101. Kuethe F, Sigusch HH, Bornstein SR, Hilbig K, Kamvissi V, Figulla HR. 1022. doi: 10.1038/nature05815.
Apoptosis in patients with dilated cardiomyopathy and diabetes: a fea- 119. Makino A, Dai A, Han Y, Youssef KD, Wang W, Donthamsetty R, Scott
ture of diabetic cardiomyopathy? Horm Metab Res. 2007;39:672–676. BT, Wang H, Dillmann WH. O-GlcNAcase overexpression reverses cor-
doi: 10.1055/s-2007-985823. onary endothelial cell dysfunction in type 1 diabetic mice. Am J Physiol
102. Yang L, Zhao D, Ren J, Yang J. Endoplasmic reticulum stress and pro- Cell Physiol. 2015;309:C593–C599. doi: 10.1152/ajpcell.00069.2015.
tein quality control in diabetic cardiomyopathy. Biochim Biophys Acta. 120. Hu Y, Belke D, Suarez J, Swanson E, Clark R, Hoshijima M, Dillmann
2015;1852:209–218. doi: 10.1016/j.bbadis.2014.05.006. WH. Adenovirus-mediated overexpression of O-GlcNAcase improves
103. Xie Z, Lau K, Eby B, Lozano P, He C, Pennington B, Li H, Rathi S, Dong contractile function in the diabetic heart. Circ Res. 2005;96:1006–1013.
Y, Tian R, Kem D, Zou MH. Improvement of cardiac functions by chron- doi: 10.1161/01.RES.0000165478.06813.58.
ic metformin treatment is associated with enhanced cardiac autophagy 121. Lehrke M, Marx N. Diabetes mellitus and heart failure. Am J Cardiol.
in diabetic OVE26 mice. Diabetes. 2011;60:1770–1778. doi: 10.2337/ 2017;120:S37–S47. doi: 10.1016/j.amjcard.2017.05.014.
db10-0351. 122. Pham D, Albuquerque Rocha N, McGuire DK, Neeland IJ. Impact of em-
104. Sandesara PB, O’Neal WT, Kelli HM, Samman-Tahhan A, Hammadah pagliflozin in patients with diabetes and heart failure. Trends Cardiovasc
M, Quyyumi AA, Sperling LS. The prognostic significance of diabetes Med. 2017;27:144–151. doi: 10.1016/j.tcm.2016.07.008.
and microvascular complications in patients with heart failure with pre- 123. von Lewinski D, Rainer PP, Gasser R, Huber MS, Khafaga M, Wilhelm
served ejection fraction. Diabetes Care. 2018;41:150–155. doi: 10.2337/ B, Haas T, Mächler H, Rössl U, Pieske B. Glucose-transporter-mediated
dc17-0755. positive inotropic effects in human myocardium of diabetic and
638  Circulation Research  February 16, 2018

nondiabetic patients. Metabolism. 2010;59:1020–1028. doi: 10.1016/j. ERK contributes to oxidative stress-induced insulin resistance in
metabol.2009.10.025. cardiac cells in vitro and in vivo. Diabetes. 2011;60:625–633. doi:
124. Sabolic I, Vrhovac I, Eror DB, Gerasimova M, Rose M, Breljak D, 10.2337/db10-1164.
Ljubojevic M, Brzica H, Sebastiani A, Thal SC, Sauvant C, Kipp H, 139. Zhang Z, Wang S, Zhou S, Yan X, Wang Y, Chen J, Mellen N, Kong M,
Vallon V, Koepsell H. Expression of Na+-D-glucose cotransporter Gu J, Tan Y, Zheng Y, Cai L. Sulforaphane prevents the development of
SGLT2 in rodents is kidney-specific and exhibits sex and species dif- cardiomyopathy in type 2 diabetic mice probably by reversing oxidative
ferences. Am J Physiol Cell Physiol. 2012;302:C1174–C1188. doi: stress-induced inhibition of LKB1/AMPK pathway. J Mol Cell Cardiol.
10.1152/ajpcell.00450.2011. 2014;77:42–52. doi: 10.1016/j.yjmcc.2014.09.022.
125. Vallon V, Rose M, Gerasimova M, Satriano J, Platt KA, Koepsell H, 140. Zhou YP, Marlen K, Palma JF, Schweitzer A, Reilly L, Gregoire FM,
Cunard R, Sharma K, Thomson SC, Rieg T. Knockout of Na-glucose Xu GG, Blume JE, Johnson JD. Overexpression of repressive cAMP
transporter SGLT2 attenuates hyperglycemia and glomerular hyperfiltra- response element modulators in high glucose and fatty acid-treated rat
tion but not kidney growth or injury in diabetes mellitus. Am J Physiol islets. A common mechanism for glucose toxicity and lipotoxicity? J Biol
Renal Physiol. 2013;304:F156–F167. doi: 10.1152/ajprenal.00409.2012. Chem. 2003;278:51316–51323. doi: 10.1074/jbc.M307972200.
126. Lytvyn Y, Bjornstad P, Udell JA, Lovshin JA, Cherney DZI. Sodium 141. Barbati SA, Colussi C, Bacci L, Aiello A, Re A, Stigliano E, Isidori AM,
glucose cotransporter-2 inhibition in heart failure: potential mecha- Grassi C, Pontecorvi A, Farsetti A, Gaetano C, Nanni S. Transcription
nisms, clinical applications, and summary of clinical trials. Circulation. factor CREM mediates high glucose response in cardiomyocytes and
2017;136:1643–1658. doi: 10.1161/CIRCULATIONAHA.117.030012. in a male mouse model of prolonged hyperglycemia. Endocrinology.
127. Raz I, Cahn A. Heart failure: SGLT2 inhibitors and heart failure – clini- 2017;158:2391–2405. doi: 10.1210/en.2016-1960.
cal implications. Nat Rev Cardiol. 2016;13:185–186. doi: 10.1038/ 142. Barwari T, Joshi A, Mayr M. MicroRNAs in cardiovascular disease. J Am
nrcardio.2016.35. Coll Cardiol. 2016;68:2577–2584. doi: 10.1016/j.jacc.2016.09.945.
128. Lei S, Li H, Xu J, Liu Y, Gao X, Wang J, Ng KF, Lau WB, Ma XL, 143. Zampetaki A, Kiechl S, Drozdov I, Willeit P, Mayr U, Prokopi M, Mayr
Rodrigues B, Irwin MG, Xia Z. Hyperglycemia-induced protein kinase A, Weger S, Oberhollenzer F, Bonora E, Shah A, Willeit J, Mayr M.
C β2 activation induces diastolic cardiac dysfunction in diabetic rats Plasma microRNA profiling reveals loss of endothelial miR-126 and
by impairing caveolin-3 expression and Akt/eNOS signaling. Diabetes. other microRNAs in type 2 diabetes. Circ Res. 2010;107:810–817. doi:
2013;62:2318–2328. doi: 10.2337/db12-1391. 10.1161/CIRCRESAHA.110.226357.
129. Li Z, Abdullah CS, Jin ZQ. Inhibition of PKC-θ preserves cardiac func- 144. Diao X, Shen E, Wang X, Hu B. Differentially expressed microRNAs
tion and reduces fibrosis in streptozotocin-induced diabetic cardiomy- and their target genes in the hearts of streptozotocin-induced diabetic
opathy. Br J Pharmacol. 2014;171:2913–2924. doi: 10.1111/bph.12621. mice. Mol Med Rep. 2011;4:633–640. doi: 10.3892/mmr.2011.489.
130. Wakasaki H, Koya D, Schoen FJ, Jirousek MR, Ways DK, Hoit BD, 145. Jordan SD, Krüger M, Willmes DM, Redemann N, Wunderlich FT,
Walsh RA, King GL. Targeted overexpression of protein kinase C beta2 Brönneke HS, Merkwirth C, Kashkar H, Olkkonen VM, Böttger T, Braun
isoform in myocardium causes cardiomyopathy. Proc Natl Acad Sci T, Seibler J, Brüning JC. Obesity-induced overexpression of miRNA-143
USA. 1997;94:9320–9325. inhibits insulin-stimulated AKT activation and impairs glucose metabo-
131. Strniskova M, Barancik M, Neckar J, Ravingerova T. Mitogen-activated lism. Nat Cell Biol. 2011;13:434–446. doi: 10.1038/ncb2211.
protein kinases in the acute diabetic myocardium. Mol Cell Biochem. 146. Trajkovski M, Hausser J, Soutschek J, Bhat B, Akin A, Zavolan M, Heim
2003;249:59–65. MH, Stoffel M. MicroRNAs 103 and 107 regulate insulin sensitivity.
132. Zhang H, Shi X, Hampong M, Blanis L, Pelech S. Stress-induced inhibi- Nature. 2011;474:649–653. doi: 10.1038/nature10112.
tion of ERK1 and ERK2 by direct interaction with p38 MAP kinase. J 147. Nair N, Kumar S, Gongora E, Gupta S. Circulating miRNA as novel
Downloaded from http://ahajournals.org by on February 10, 2022

Biol Chem. 2001;276:6905–6908. doi: 10.1074/jbc.C000917200. markers for diastolic dysfunction. Mol Cell Biochem. 2013;376:33–40.
133. Wang Y, Zhou S, Sun W, McClung K, Pan Y, Liang G, Tan Y, Zhao Y, doi: 10.1007/s11010-012-1546-x.
Liu Q, Sun J, Cai L. Inhibition of JNK by novel curcumin analog C66 148. Liu X, Liu S. Role of microRNAs in the pathogenesis of diabetic cardio-
prevents diabetic cardiomyopathy with a preservation of cardiac metallo- myopathy. Biomed Rep. 2017;6:140–145. doi: 10.3892/br.2017.841.
thionein expression. Am J Physiol Endocrinol Metab. 2014;306:E1239– 149. Garcia NA, Moncayo-Arlandi J, Sepulveda P, Diez-Juan A.

E1247. doi: 10.1152/ajpendo.00629.2013. Cardiomyocyte exosomes regulate glycolytic flux in endothelium by di-
134. Gurusamy N, Watanabe K, Ma M, Zhang S, Muslin AJ, Kodama M, rect transfer of GLUT transporters and glycolytic enzymes. Cardiovasc
Aizawa Y. Dominant negative 14-3-3 promotes cardiomyocyte apopto- Res. 2016;109:397–408. doi: 10.1093/cvr/cvv260.
sis in early stage of type I diabetes mellitus through activation of JNK. 150. Wang X, Huang W, Liu G, Cai W, Millard RW, Wang Y, Chang J, Peng
Biochem Biophys Res Commun. 2004;320:773–780. doi: 10.1016/j. T, Fan GC. Cardiomyocytes mediate anti-angiogenesis in type 2 diabetic
bbrc.2004.06.023. rats through the exosomal transfer of miR-320 into endothelial cells.
135. Yang J, Park Y, Zhang H, Xu X, Laine GA, Dellsperger KC, Zhang J Mol Cell Cardiol. 2014;74:139–150. doi: 10.1016/j.yjmcc.2014.05.001.
C. Feed-forward signaling of TNF-alpha and NF-kappaB via IKK- 151. Wang X, Gu H, Huang W, Peng J, Li Y, Yang L, Qin D, Essandoh K,
beta pathway contributes to insulin resistance and coronary arteriolar Wang Y, Peng T, Fan GC. Hsp20-mediated activation of exosome biogen-
dysfunction in type 2 diabetic mice. Am J Physiol Heart Circ Physiol. esis in cardiomyocytes improves cardiac function and angiogenesis in
2009;296:H1850–H1858. doi: 10.1152/ajpheart.01199.2008. diabetic mice. Diabetes. 2016;65:3111–3128. doi: 10.2337/db15-1563.
136. Mariappan N, Elks CM, Sriramula S, Guggilam A, Liu Z, Borkhsenious 152. Young LH, Wackers FJ, Chyun DA, Davey JA, Barrett EJ, Taillefer R,
O, Francis J. NF-kappaB-induced oxidative stress contributes to mito- Heller GV, Iskandrian AE, Wittlin SD, Filipchuk N, Ratner RE, Inzucchi
chondrial and cardiac dysfunction in type II diabetes. Cardiovasc Res. SE; DIAD Investigators. Cardiac outcomes after screening for asymp-
2010;85:473–483. doi: 10.1093/cvr/cvp305. tomatic coronary artery disease in patients with type 2 diabetes: the
137. Niture SK, Khatri R, Jaiswal AK. Regulation of Nrf2-an update. Free DIAD study: a randomized controlled trial. JAMA. 2009;301:1547–1555.
Radic Biol Med. 2014;66:36–44. doi: 10.1016/j.freeradbiomed.2013. doi: 10.1001/jama.2009.476.
02.008. 153. Fang ZY, Schull-Meade R, Leano R, Mottram PM, Prins JB, Marwick
138. Tan Y, Ichikawa T, Li J, Si Q, Yang H, Chen X, Goldblatt CS, Meyer TH. Screening for heart disease in diabetic subjects. Am Heart J.
CJ, Li X, Cai L, Cui T. Diabetic downregulation of Nrf2 activity via 2005;149:349–354. doi: 10.1016/j.ahj.2004.06.021.

You might also like