You are on page 1of 19

Pflügers Archiv - European Journal of Physiology

https://doi.org/10.1007/s00424-021-02651-x

INVITED REVIEW

Understanding diabetes‑induced cardiomyopathy


from the perspective of renin angiotensin aldosterone system
Vijayakumar Sukumaran1 · Narasimman Gurusamy2 · Huseyin C. Yalcin1 · Sundararajan Venkatesh3

Received: 19 May 2021 / Revised: 2 December 2021 / Accepted: 3 December 2021


© The Author(s), under exclusive licence to Springer-Verlag GmbH Germany, part of Springer Nature 2021

Abstract
Experimental and clinical evidence suggests that diabetic subjects are predisposed to a distinct cardiovascular dysfunction,
known as diabetic cardiomyopathy (DCM), which could be an autonomous disease independent of concomitant micro and
macrovascular disorders. DCM is one of the prominent causes of global morbidity and mortality and is on a rising trend
with the increase in the prevalence of diabetes mellitus (DM). DCM is characterized by an early left ventricle diastolic
dysfunction associated with the slow progression of cardiomyocyte hypertrophy leading to heart failure, which still has no
effective therapy. Although the well-known “Renin Angiotensin Aldosterone System (RAAS)” inhibition is considered a
gold-standard treatment in heart failure, its role in DCM is still unclear. At the cellular level of DCM, RAAS induces vari-
ous secondary mechanisms, adding complications to poor prognosis and treatment of DCM. This review highlights the
importance of RAAS signaling and its major secondary mechanisms involving inflammation, oxidative stress, mitochondrial
dysfunction, and autophagy, their role in establishing DCM. In addition, studies lacking in the specific area of DCM are also
highlighted. Therefore, understanding the complex role of RAAS in DCM may lead to the identification of better prognosis
and therapeutic strategies in treating DCM.

Keywords  Diabetic cardiomyopathy · Rennin angiotensin aldosterone system · Oxidative stress · Inflammation ·
Mitochondrial dysfunction · Autophagy

Introduction that directly induces abnormalities and alterations in cardiac


muscle and coronary function [100]. Such dysfunction leads
Cardiovascular disease (CVD) is the leading cause of death to impaired cardiac contractility and ventricular compliance
worldwide, where diabetes mellitus (DM)–related cardiac resulting in “diabetic cardiomyopathy” (DCM). DCM could
abnormalities increase morbidity and mortality further when be developed independently of coronary artery disease and
compared to CVD alone [108, 115]. DM is a chronic disease hypertension that often accompanies DM and is induced by
currently affecting approximately 415 million people glob- a range of systemic and localized metabolic changes result-
ally and is estimated to rise to 642 million by 2040 [64]. Dia- ing in structural and functional cardiac abnormalities [49,
betic subjects are highly prone to develop CVD due to insu- 189]. Therefore, DCM could be an autonomous disease inde-
lin resistance, hyperglycemia, and hyperlipidemia, which pendent of other cardiovascular manifestations. DCM is still
play an essential pathophysiological role in developing DM a significant challenge despite the availability of advanced
therapies to treat heart disease. The lack of knowledge in
the molecular mechanisms of the disease is one of the rea-
* Vijayakumar Sukumaran
vijay@qu.edu.qa; svkumar79@gmail.com sons for the challenge, even though various factors are being
implicated and studied.
1
Biomedical Research Center, Qatar University, Al‑Tarfa, The well-known Framingham Heart Study showed that
2371 Doha, Qatar DCM is one of the highest contributors to mortality among
2
Department of Bioscience Research, University of Tennessee the comorbidities that contribute to heart failure [31, 56].
Health Science Center, Memphis, TN, USA DM is a strong and independent risk factor for developing
3
Department of Microbiology, Biochemistry and Molecular heart failure, and DCM is reported in 16–40% of type 2 DM
Genetics, Rutgers-New Jersey Medical School, Newark, NJ, cases [117]. Nonetheless, type 1 DM and type 2 DM differ
USA

13
Vol.:(0123456789)
Pflügers Archiv - European Journal of Physiology

in their etiology and metabolic profiles even though both pri- blood pressure drop [28]. A fall in blood pressure activates
marily result in similar characteristics of DCM, which devel- sympathetic nervous system (SNS) that indirectly releases
oped from a multi-factorial consequence. Many molecular renin through b1 receptor stimulation in juxtaglomerular
mechanisms have been proposed to contribute to the patho- cells. The released renin cleaves the hepatic angiotensino-
genesis of DCM, but the complex interactions between many gen into angiotensin I (ANG-I), which is then converted
of the factors involved remain to be elucidated [14, 111]. into ANG-II by a pulmonary angiotensin-converting
The renin–angiotensin–aldosterone system (RAAS) is one enzyme (ACE). The ANG-II restores the blood pressure
of the major signaling pathways involved in developing car- by its various physiological actions such as vasoconstric-
diac abnormalities [8]. However, its role and molecular mech- tion, renal sodium and water retention, stimulating the
anism that contributes to the development of DCM are poorly sympathetic nervous system, and aldosterone secretion
understood. For better prevention and strategies to improve in the adrenal glands [177]. RAAS is composed of two
the treatment of DCM, it is an unmet need to understand the opposing axes (Fig. 1). The first axis is driven by the ACE,
molecular mechanisms and the pathophysiology of DCM. which produces the end product ANG-II. The A ­ T1 recep-
Therefore, in this review, we focus on RAAS and its associa- tor is the main effector of ANG-II, which mediates most
tion with various factors such as oxidative stress, inflamma- of its dominant biological actions. In contrast, the A ­ T2
tion, autophagy, and mitochondrial dysfunction, and how it receptor has been shown to oppose many of the activities
influences these factors in contributing to DCM development. of the A­ T 1 receptor in the ACE/ANG-II axis [204] and
mediates at least in part some of the beneficial effects of
the ­AT1 receptor blockade on the blood vessels, heart, and
Pathophysiology of DCM kidneys [90, 120]. Unlike A ­ T1, the role of A
­ T2 receptors
is not clearly understood. However, it is not completely
Unlike cardiomyopathy, the pathophysiology of DCM is com- clear how diabetes affects the expression and function of
plicated because of multiple factors involved in its development these receptors.
over time since from the initial stage of hyperglycemia. DCM The second RAAS axis involves ACE-2-mediated
can be characterized by an initial left ventricular (LV) diastolic hydrolysis of ANG-II, leading to the production of ANG
dysfunction with preserved LV ejection fraction (LVEF) that 1–7, which binds to the Mas receptor as the primary effec-
occurs significantly before changes in systolic function that are tor. This axis mediates the vasodilator, anti-inflammatory,
detectable by routine assessment with echocardiography [49, anti-oxidative, and anti-fibrotic actions that oppose ­AT1
186]. This diastolic dysfunction is detectable even without LV receptor-mediated effects. However, the available evidence
hypertrophy, suggesting that the LV dysfunction could be inde- suggests that in DM, the balance of RAAS activation is
pendent of LV hypertrophy and caused by diabetes alone [3, primarily shifted towards the activation of the ACE/ANG-
66, 157]. However, systolic dysfunction has also been reported II/AT1 receptor pathway [54]. Supporting evidence also
in diabetes but at a lower rate than diastolic dysfunction. Sus- shows that hyperglycemia and DM-induced cardiac dys-
tained hyperglycemia is undoubtedly one of the main pathogenic function can be reversed by pharmacological inhibition of
factors that cause structural and functional abnormalities at the the ­AT1 receptor pathway [142]. Interestingly, Alaman-
cardiomyocyte level [69]. The changes in the myocardium also dine, a newly identified heptapeptide generated by the
depend on the duration of diabetes. Ultimately, fibrosis at the catalytic action of ACE-2 angiotensin A or directly from
end may reduce the left ventricular function [62, 102]. During ANG 1–7 has been identified as another element in the
the development of DCM, insulin resistance, impaired calcium protective ACE/ANG-II receptor axis [93]. Alamandine
homeostasis, metabolic alterations including changes in sub- is reported to act by binding to Mas-related G-protein-
strate utilization such as increased fatty acid oxidation, inhibi- coupled receptor (MrgD) and is potentially antagonized
tion of glucose oxidation, oxidative stress, inflammation, and by PD-123319, an A ­ T2, and MrgD antagonist but not by
the aberrant activation of RAAS signaling may directly impair the Mas receptor antagonist (A-779) [190]. However, it has
cardiomyocyte function [13, 14, 191]. Importantly, these key been shown that MrgD is a second receptor for ANG 1–7
contributors increase myocardial fibrosis and cellular death [181]. But further studies are warranted to confirm these
resulting in cardiomyopathy followed by heart failure [13, 14]. findings and investigate how alamandine affects the struc-
ture, function of the heart, and morbidity in the long term.
Activation of RAAS elements participates in the long-
RAAS in the development of DCM term regulation of cardiovascular homeostasis, including
modulation of cardiac contractility and coronary blood
The main effector of RAAS is angiotensin II (ANG-II). flow via sustained activation of local ANG-II [35, 217].
On the other hand, renin responds to low sodium concen- Though RAAS is considered a life-saving mechanism by
tration at the macula densa cells, not by a direct effect of regulating blood pressure during hemorrhage, RAAS also

13
Pflügers Archiv - European Journal of Physiology

Fig. 1  Opposing arms of RAAS involved in DCM

increases blood pressure when activated inappropriately, cardiovascular morbidities confirming their role in DCM.
leading to chronic hypertension that can contribute to the Importantly, it has been demonstrated that activating RAAS
development of cardiovascular diseases [206]. Beyond this, signaling in diabetes results in tissue-specific increases in
various clinical trials have shown that inhibiting RAAS by ANG-II, the primary physiological effector molecule [96].
ACE inhibitors significantly reduced mortality in patients ANG-II contributes to numerous alterations in DCM, includ-
suffering from heart failure, suggesting that RAAS could ing endothelial dysfunction, cardiac hypertrophy, and deple-
also affect the heart apart from blood vessels [53]. Further tion of tissue antioxidant levels, which resulted in increased
studies have shown that RAAS could promote ventricular oxidative stress and vascular inflammation [45, 46]. Fur-
hypertrophy, fibrosis, and myocardial infarction apart from ther, ANG-II actions are associated with endothelial dys-
affecting blood pressure [27, 103]. Besides, recent studies function, resulting in part from decreased nitric oxide (NO)
have also shown the existence of tissue-specific RAAS, availability in endothelial cells [155]. Conversely, attenuat-
which not only play a role in the pathophysiology of the ing ANG-II improved endothelial function by suppressing
development of CVD but also implicated in the develop- reactive oxygen species (ROS) production and restoring NO
ment of diabetic cardiovascular abnormalities [211], where availability [127]. Further, ACE inhibition has been shown
it is involved in cardiomyocyte death, interstitial fibrosis, to improve mortality and morbidity in diabetic patients by
hypertrophy, and atherosclerosis [19, 52, 109]. Therefore, decreasing cardiovascular complications [70, 218]. The roles
inhibiting the RAAS pathway by ACE inhibitors and ANG- of RAAS activation and the role of two axes in DCM devel-
II receptor blockers showed a promising outcome in diabetic opment are discussed further in detail.

13
Pflügers Archiv - European Journal of Physiology

ACE/ANG‑II/AT1 and ­AT2 receptor pathways Nonetheless, although ARB and ACEi have been shown
in DCM to reduce pathological remodeling, cardiovascular events,
and mortality in diabetic patients, these conventional
In the classical RAAS axis, ANG-II is produced from ANG-I therapies that target only one arm of the RAAS do not
by the action of ACE. ANG-II is a well-known bioactive achieve a high degree of efficacy (Table 1). In general,
peptide involved in the exaggeration of cardiovascular disease cardiovascular mortality is reduced by 20–40% with either
[45]. At the onset of DM and in other metabolic disorders, conventional therapies, including either ARB or ACEi [30,
activation of ACE/ANG-II/AT 1 receptor axis causes 73, 98]. Importantly, a combination of ARB or ACEi with
maladaptive effects, including vasoconstriction, inflammation, the renin inhibitor Aliskiren did not improve cardiorenal
fibrosis, apoptosis, cellular growth and migration, and fluid endpoints in diabetic patients with renal impairment [133].
retention [87, 110]. ­AT1 receptors are distributed throughout Hence, these findings have been part of the motivation for
most organs, whereas ­AT2-receptor expression is observed investigating the anti-inflammatory actions of stimulating
only in a few organs after birth and is upregulated in some the non-classical arm of RAAS. In a most recent study,
pathological states [32]. Chronic A ­ T1-receptor stimulation it has been shown for the first time that activation of the
is known to cause hypertension, stroke, cardiovascular ­AT2 receptor opposes ANG-II-mediated atherosclerosis
events, and renal diseases. It has also been reported that the via ­AT1 receptor [24]. Treatment with a Compound 21, a
blockade of A­ T1 receptor promotes longevity [11]. Increased selective ­AT2 agonist, potently ameliorated aortic plaque
activation of the ACE/ANG-II/AT1R pathway in rats with deposition in an in vivo animal model of diabetes-associ-
enhanced tissue renin-ANG expression was observed after ated atherosclerosis through the suppression of inflamma-
6 weeks of STZ-induced diabetes. This also resulted in an tory mediators and oxidative stress [24]. Similarly, activa-
impairment of both active and passive phases of diastolic LV tion of AT2 also ameliorated myocardial hypertrophy in
function accompanied by interstitial fibrosis, cardiac myocyte diabetic hearts [20]. Therefore, the activation or balance
hypertrophy, and increased apoptosis along with an increase between ­AT1 and A ­ T2 receptor signaling plays an impor-
in transforming growth factor-beta (TGF-β) activity [25]. The tant role in DCM development, but how the signaling is
modulation of ­AT1R/ERK/MAPK signaling plays a crucial regulated remains poorly understood.
role in developing accelerated diabetes-induced cardiac
dysfunction in DCM [92]. Regimes that reduce activation of
the ACE/ANG-II/AT1 receptor pathway can slow down the ACE‑2/ANG 1–7/Mas receptor pathway
progression of DCM partially. We have previously shown that in DCM
­AT1 receptor antagonists such as olmesartan and telmisartan
attenuated apoptosis, oxidative stress, endoplasmic reticulum The relatively recent discovery of new family members
stress, and cardiac inflammatory mediators and significantly of the RAAS system, such as ACE-2, ANG 1–7, and
improve myocardial function [172, 173]. These findings ANG 1–9, has spurred new interest in understanding the
provide the molecular basis for the increased benefits relationship between the RAAS signaling pathways and
observed with both A ­ T1 receptor blockade (ARB) and ACE cardiovascular disease. ACE-2, a resemblance of ACE,
inhibitors (ACEi) in ameliorating DCM in diabetic patients catalyzes the conversion of ANG-II to a vasodilator hep-
[123, 176]. tapeptide ANG 1–7, whereas ANG-I is converted into the

Table 1  ACE/ANG-II type 1 receptor axis modulation during the development of DCM


Organ/model Approach Effect in the heart References

Isolated cardiomyocytes AT1 antagonist ↓ NADPH oxidase, ↓ ROS production Privratsky et al. 2003 [142]
Isolated cardiomyocytes Candesartan ↓ Oxidative stress, ↓ PKC phosphorylation Yaras et al. 2007 [203]
Heart Irbesartan (IRB) ↓ Cardiac inflammation, fibrosis Westermann et al. 2007 [196]
Heart Candesartan ↓ Oxidative stress, fibrosis, myocyte apoptosis Singh et al. 2008 [164]
Isolated cardiomyocytes AT1R-KO mice, Valsartan ↓ Reactive oxygen species, cardiac apoptosis Yong et al. 2013 [208] 
Heart Valsartan ↓ Oxidative stress, cardiomyocyte apoptosis Zhang et al. 2013 [212]
Heart Valsartan ↓ Reactive oxygen species, fibrosis Thomas et al. 2013 [183]
Heart AT1R-KO mice ↓ NADPH oxidase Nagotomo et al. 2014 [121]
Eplerenone ↓ Apoptosis
Heart losartan ↓ oxidative stress and cardiac apoptosis Hussien et al. 2015 [71]
Heart Irbesartan ↓ oxidative stress and fibrosis, inflammation Watanabe et al. 2016 [193]
Heart Compound 21 + losartan ↓ oxidative stress and myocardial hypertrophy Castoldi et al. 2019 [20]

13
Pflügers Archiv - European Journal of Physiology

inactive nonapeptide ANG 1–9; thereby, ACE-2 effectively vasculature through a telomerase-dependent manner in
functions as an endogenous ACE inhibitor [154]. The patients with coronary artery disease [44]. While directly
mechanisms underlying the beneficial actions of ACE-2 targeting the A­ T1 receptor pathway failed to improve the
and ANG 1–7 in glucose metabolism are thought to be outcome of cardiovascular patients [202], targeting the coun-
multifaceted. First, increased degradation of ANG-II by ter-regulatory ACE-2/Mas receptor axis by enhancing ANG
ACE-2 might be important in regulating glucose metabo- 1–7 actions may yet represent a novel therapy for DCM, for
lism [77]. Second, ANG 1–7 can activate the phosphati- which there are limited studies (Table 2).
dylinositol-3-kinase–Akt pathway [40, 60, 118, 152, 182] It is widely recognized that the increment in oxidative
and thereby facilitate insulin-mediated signaling and glu- stress is an important cause of disruption in cell–cell com-
cose uptake. ANG 1–7 might also normalize the defective munication as a consequence of ANG-II and aldosterone
insulin signaling induced by ANG-II by preventing the formation and activation of the ACE/ANG-II/AT1 receptor
serine phosphorylation of insulin receptor substrate [143, pathway [36]. Our previous study showed that ANG 1–7
179], probably via inhibition of the activation of MAPK elicited by the activation of ACE-2 has beneficial effects in
pathways induced by ANG-II [6, 213]. Moreover, it is the heart [171], counteracting the harmful effects of elevated
now suggested that ANG 1–7 facilitates glucose uptake ANG-II [92], and re-establishing impulse conductance dur-
by the upregulation of glucose transporter type (GLUT) ing myocardial ischemia [33]. Hence, ANG 1–7 might play
4, which is the major glucose transporter in humans [178]. a role in maintaining functional cellular connections in the
Although some studies have shown that the counter-reg- heart by enhancing gap junction permeability with conse-
ulatory ACE2/ANG 1–7/Mas axis may promote inflam- quent improvement of metabolic interactions between car-
matory and oxidative stress responses, recent evidence diac cells. Recently, ANG 1–7 was also shown to increase
demonstrates that ANG 1–7 negatively modulates inflam- the gap junction permeability by activating protein kinase
mation, NADPH oxidase, fibrogenesis, and apoptotic A and the phosphorylation of gap junction channels [34].
pathways in DCM [151]. In addition, it has been recently Further, non-peptide ANG 1–7 receptor agonist AVE0991
demonstrated that ACE-2 also negatively regulates inflam- has been shown to rescue hyperglycemia-induced pathologi-
mation, insulin resistance, and cardiac metabolism altera- cal changes in the heart [46]. Various other studies have also
tions in obesity-mediated cardiac dysfunction [135]. pointed out the therapeutic benefits of ANG 1–7 in reduc-
Furthermore, others have reported that Irbesartan ame- ing or preventing cardiovascular abnormalities caused by
liorates LV remodeling after myocardial infarction in part diabetes through multiple mechanisms such as reducing
as a result of ACE-2 upregulation [84]. Chronic ANG 1–7 lipid accumulation, systemic fat mass, inflammation, and
treatment significantly restored nitric oxide levels in the increased insulin-stimulation [68, 162]. Thus, the balance of

Table 2  ACE-2/ANG-(1–7) Mas receptor axis modulation during the development of DCM


Organ/model Approach Effect in the heart References

Heart ACE-2 deletion ↑ Ventricular dilation Oudit et al. 2007


↓ Intrinsic myocardial contractility [130]
Heart: cardiac fibrosis ANG 1–7 ↓ Hypertrophy, Interstitial fibrosis Grobe et al. 2007 [63]
Isolated perfused hearts ANG 1–7 ↑ Vascular response to ANG-II, endothelin-1 Benter et al. 2007 [12]
Heart ANG 1–7 ↓ LV protein and collagen content Singh et al. 2011
[162]
Heart ANG 1–7 ↓ Superoxide levels, NADPH activity Mori et al. 2014 [113]
↑ Insulin sensitivity
↓ Fibrosis, inflammatory cytokines
Isolated cardiomyocytes ANG 1–7 ↑ Insulin sensitivity Santos 2014 [153]
Heart ANG 1–7 ↓ Right ventricle fibrosis and apoptosis Hao et al. 2015 [68]
Heart ANG 1–7 ↓ Fibrosis, apoptosis, Superoxide levels Hao et al. 2015 [68]
↓ SERCA-2, Collagen-3, TGF-β1
Heart ANG 1–7 ↓ Fibrosis, hypertrophy and apoptosis Papinska et al. 2016
↓ Inflammatory cytokines, oxidative stress [132]
Heart ANG 1–7 ↓ Fibrosis, hypertrophy and apoptosis Sukumaran et al. 2017
[170]

Benter et al., 2007; Grobe et al., 2007; Oudit et al., 2007; Singh et al., 2011; Mori et al., 2014; Santos et al., 2014; Hao et al., 2015; Papinska et
al., 2016; Sukumaran et al., 2017

13
Pflügers Archiv - European Journal of Physiology

activation between the two RAAS axes may play an essential subsequent report in nondiabetic mice and shown that block-
role in the maintenance of cellular connections in the myo- ade of A­ T1 receptors by Irbesartan or infusion of ANG 1–7
cardium by regulating gap junction permeability between exerted comparable levels of anti-inflammatory effects in
the cardiac cells. Still, the loss of counterbalance during the ACE-2 null hearts of pressure-overloaded mice [134].
the development of DCM could trigger the progression of On the other hand, there is little direct evidence that sys-
the disease. temic inflammation markers such as c reactive protein are
frequently reported to be elevated in DM [91]. Some of the
interventions that have demonstrated protective effects are
RAAS and inflammation in DCM likely to be a consequence of a reduction in cardiac inflam-
mation through various mechanisms involving ­AT1R antago-
RAAS activation appears to play a critical role in the ini- nists and inhibition of both the MAPK pathways and inter-
tiation and potentiation of cardiac inflammation in DCM. leukin-converting enzyme [196, 197]. Indeed, resveratrol
It is known that the chronic inflammation induced by pro- has been shown to attenuate cardiac dysfunction and inflam-
inflammatory cytokines contributes to DCM development mation through activation of ­AT1R/ERK/p38MAPK signal-
[39, 67, 86]. Serum levels of inflammatory cytokines such ing pathway in STZ-induced diabetic rats [59]. Therefore,
as interleukin (IL)-6, IL-1β, and tumor necrosis factor- independent of RAAS signaling, RAAS-induced inflamma-
alpha (TNF-α) are increased in diabetic patients [41]. These tion may play an additive role in DCM development. Further
increased inflammatory cytokines cause insulin resistance by studies on how RAAS induces the activation of inflamma-
decreasing insulin signaling due to reduced IRS-1 tyrosine tory cytokines in the heart are needed to target the RAAS-
phosphorylation and the activation of PI3K and Akt [74]. In inflammatory pathways. Therefore, it is essential to differ-
addition, inflammatory cytokines could also directly affect entiate between RAAS-induced and RAAS-independent
the beta cells, leading to their death and reduction in the inflammation in DCM development to specifically target
numbers [42]. Supporting data show that these inflammatory the inflammation source.
cytokines are found in the hearts of diabetic patients [210].
These inflammatory mediators have been shown to promote
myocardial dysfunction leading to heart failure [197]. For RAAS and oxidative stress in DCM
example, by overexpressing TNF-α, one of the inflamma-
tory cytokines increased cardiac fibrosis, and hypertrophy, Although inflammation partly contributes to cardiac remod-
leading to LV dysfunction [174, 207]. Also, activation of the eling during DCM, the presence of oxidative stress during
pro-inflammatory transcription factor nuclear factor kappa B inflammation is inevitable, and OS is also one of the most
(NF-κB) has been shown to trigger the generation of ROS/ commonly implicated reasons in various diseases, including
reactive nitrogen species (RNS) in diabetic hearts, isolated DCM [85]. Oxidative stress appears to be an early contribu-
human cardiomyocytes, and coronary artery endothelial cells tor to the development and progress of DM, and its associ-
when exposed to high glucose concentration [107, 145]. ated complications could be dependent or independent of
Though initially it is not clear that the inflammation is inflammation. A recent study shows that insulin resistance
a direct effect of RAAS activation or as a result of indirect leads to enhanced ROS production, inducing myocardial
consequences, later studies have demonstrated that block- oxidative stress and diminution of the antioxidant defense
ade of ANG-II signaling significantly reduces the levels of mechanisms in the heart [189]. The oxidative damage caused
pro-inflammatory mediators and oxidative stress in various by ROS from a hyperglycemic state plays a vital role in dia-
models of cardiac inflammation [124, 172, 173, 198]. This betic injury in multiple cell types, including the myocardium
is further confirmed by a recent study showing that inhibit- [65, 159, 160, 180]. In addition, ROS promotes a vicious
ing RAAS by either Losartan or Perindopril reduced the cycle of oxidative stress leading to changes in gene expres-
systemic and regional inflammatory release of monocytes sion, faulty signal transduction, cardiomyocytes apoptosis,
and neutrophils which resulted in the prevention of cardiac and stimulation of connective tissue growth factor, fibro-
rupture during acute myocardial infarction [58]. On the other sis, and the formation of AGEs, which further exacerbate
hand, chronic treatment with ANG 1–7 completely rescued the stiffening of diabetic hearts [106, 163]. Experimental
diastolic dysfunction in the diabetic (db/db) mouse model studies have consistently been shown that oxidative stress is
by suppressing inflammation [113, 131]. These evidences involved in vascular remodeling and diastolic dysfunction
strongly supports that independent of RAAS-induced car- by activating many signaling pathways, including MAPK,
diac modeling, induction of inflammatory response may calcium channels, and redox-sensitive transcription factors.
be additional factors affecting cardiac function. Moreover, Activation of these signaling pathways causes vascular
critical roles for the increased action of ANG-II and/or loss injury through apoptosis in the vessel wall, DNA damage,
of ANG 1–7-mediated effects have been established in a and by evoking the expression of proinflammatory genes

13
Pflügers Archiv - European Journal of Physiology

Fig. 2  The source of fibroblasts


in the fibrotic myocardium.
Resident cardiac fibroblasts can
be activated for its proliferation
or transdifferentiation by ANG-
II, ROS, and TGF-β. Addition-
ally, damaged cardiac myocytes,
endothelial cells, and pericytes
can be transdifferentiated into
fibroblasts or myofibroblasts

[112, 119, 185]. Notably, the elevated ROS levels and com- ANG-II [80]. Such reduction has also been accomplished
promised antioxidant mechanisms are associated with dias- by ­AT1 blocker losartan and the ­O2 scavenger Tiron sug-
tolic dysfunction and cardiac remodeling in streptozotocin gesting the potential role of RAAS in inducing oxidative
(STZ)-induced DM, as well as genetic models of type 2 DM stress in the development of DCM [80]. On the other hand,
[72, 165, 166]. In addition, oxidative stress also appears to metallothionein, a group of small cysteine-rich metal-bind-
be an essential driver in the pathogenesis of fibrosis in DCM ing proteins, possesses antioxidant activity, and suppresses
[165, 166, 184]. On the other hand, other cell types in the ANG-II-dependent NOX-induced nitrosative damage and
heart including cardiac myoctes could be prone to transdif- cell death in diabetic hearts [216].
ferentiation into fibroblasts (Fig. 2). Evidence shows that Although there is enough evidence that RAAS directly
inhibition of NADPH oxidase in young rats shortly after the influences OS in DCM, studies also show that RAAS might
onset of DM attenuates subsequent interstitial fibrosis in the indirectly establish oxidative stress in the myocardium dur-
myocardium and completely prevents systolic and diastolic ing DM. For example, hypertension that affects the diabetic
function in diabetic rats [104]. The pro-inflammatory-medi- heart could be induced by an increase in the expression
ated ROS contribution is also evident from increased expres- of the ­AT1 receptor and ANG-II binding to ­AT1 receptor
sion of TNF-α, NF-κB, and collagen in the heart of rats with through treatment with one of the oxidants, l-buthionine
DM, which was significantly diminished by an antioxidant sulfoximine (BSO), that positively correlates oxidative stress
N-acetyl cysteine [95, 200]. Further, hyperglycemia-induced and RAAS [9]. This is possibly by ­AT1-induced phosphoryl-
inflammation and fibrosis were also ameliorated by treat- ation of Jak kinase-2, which forms a molecular complex with
ment with an antioxidant during the development of DCM calmodulin that interacts with NHE3, resulting in increased
[172], which also implies the inflammation and oxidative sodium reabsorption and blood pressure. Further, treating
stress are interconnected [166, 172]. with antioxidant tempol or ­AT1 receptor blocker Candesar-
Evidence that ANG-II promotes OS in DCM progres- tan prevents oxidative stress–induced pathological changes
sion has also been demonstrated when insulin-like growth in ­AT1-induced hypertension, suggesting the indirect role of
factor-1(IGF-1) overexpression reduces OS and myocyte RAAS-induced OS in the heart [10]. Also, additional stimu-
apoptosis in the diabetic heart [80]. These are accompanied lation of other pathways involving NF-Kβ promotes inflam-
by a reduction in nitrotyrosine, ­H2O2, and ·OH reactive oxy- mation and oxidative stress resulting in the aggravation of
gen radicals in myocytes, probably by the decrease in the myocardial injury since RAAS is also shown to be localized
expression of p53, Bax, angiotensinogen, ­AT1 receptors, and in many tissues, including the heart [10].

13
Pflügers Archiv - European Journal of Physiology

Established oxidative stress may further activate the are interconnected and it has been shown that in diabetes
RAAS and proceed as a vicious cycle in establishing DCM. RAAS hyperactivation is associated with ROS generation,
Therefore, treating with antioxidants alone may not be effec- resulting in cell damage. RAAS and mitochondria have a
tive but could be an added strategy in treating DCM. Sup- mutual role in the production of cellular ROS. While ANG-
porting experimental studies have shown that antioxidant II activates NADPH oxidase and assists eNOS uncoupling,
therapies can at least partially normalize the increased ROS it also mediates mtROS generation, which further impairs
production and consequently reduce the severity of DM mitochondrial energy production [43, 88]. Increased expres-
[165, 166]. Notably, a high plasma glucose concentration sion of ANG-II via activation of ­AT1R is shown to cause
induces an imbalance between ROS species and NO bio- morphological and functional impairment of mitochondria
availability as an early event in the progression of DM that in cardiomyocytes. In addition, it has also been shown that
causes endothelial dysfunction [138]. Many of the antioxi- mineralocorticoids can activate the fibrotic effects of ­AT1R
dant treatment clinical trials have not completely rescued signaling by increasing the ANG-II-induced cardiac oxida-
the cardiovascular abnormalities [26, 146, 165, 166, 205]. tive stress (Fig. 2) [175, 214].
Therefore, oxidative stress may not be the sole cause of Further evidence shows that overexpressing Ren2 chronically
DCM but could be a mediator in DCM development. Added activates the tissue RAAS causing mitochondrial interdigitat-
antioxidant therapy may be beneficial if the source of oxida- ing accompanied by hypertension, diabetes, and cardiovascular
tive stress is correctly identified. damage [167]. However, inhibition of mineralocorticoid recep-
tors in this Ren2 transgenic mouse has been shown to rescue
mitochondrial abnormalities, suggesting that RAAS could
RAAS and its role in mitochondrial induce DCM phenotype possibly through mitochondrial dys-
dysfunction and contribution to DCM function [167]. Another evidence suggesting that RAAS con-
tributes to mitochondrial dysfunction in the heart is that Ren2
Mitochondria are the central stages for metabolism and are overexpressed mice-induced aortic remodeling was rescued
highly prone to diabetes-induced changes. Mitochondria by treating with mitochondrial specific antioxidant tempol,
regulate most of the critical events such as ATP although NADPH oxidase is shown to contribute to the majority
production, calcium handling, β-fatty acid oxidation, of ROS generation in the Ren2 transgenic mouse [195]. There-
and mitophagy in the cell. Therefore, mitochondrial fore, detailed studies are warranted to confirm the contribution
dysfunction generally has var ious pathological of RAAS-induced mitochondrial dysfunction in DCM.
consequences that affect most of these key regulatory
pathways and trigger cardiac remodeling. It has been
shown that a severe reduction in mitochondrial ATP RAAS and autophagy in DCM
production was observed in type 2 DM, which was
associated with cardiac abnormalities such as diastolic Autophagy is a highly conserved homeostatic process that
dysfunction [114, 137]. Also, type 2 DM patients plays an essential role in the heart to maintain normal home-
undergoing coronary artery bypass surgery showed ostasis by recycling long-lived molecules, including cellular
increased oxidative stress associated with reduced organelles, by targeting them into lysosomes for degradation.
respiration in their left arterial appendage tissue [5]. Therefore, autophagy could be a cardioprotective process by
In addition, change in mitochondrial energy was also reducing cellular damage to the heart against various stress
observed during impaired cardiac insulin signaling in conditions [1, 122, 150, 158]. Also, autophagy can be det-
cardiac-specific insulin receptor deleted mice [15]. rimental, depending on the extent of autophagy and experi-
These shreds of evidence strongly suggest that cardiac mental animal models in question. Defective autophagy is
mitochondrial dysfunction has been associated with the already shown to influence skeletal and cardiac dysfunction
development of DCM independent of RAAS, including [55, 125]. The very first and early studies in 2009 demon-
through O-GlcNAcylation modification [16, 75, 76, strated that RAAS influences autophagy in cardiomyocytes,
105, 144]. Although RAAS could directly induce where ANG-II increases autophagosomes in the presence of
mitochondrial dysfunction in the heart by decreasing ­AT1 receptor overexpression. However, these responses were
mitochondrial enzyme and ETC complex activities, its inhibited by co-expressing a high A ­ T2/AT1 receptors ratio,
contribution to DCM through mitochondrial dysfunction suggesting a potential reciprocal regulation of autophagy
is unknown [78]. by ­AT1 and A
­ T2 receptors [140, 141]. However, in contrast,
Emerging studies suggest the participation of RAAS ANG 1–7 is negatively associated with autophagy but pro-
in mitochondrial dysfunction contributing to DCM [167, vides a protective function by reducing oxidative stress and
195]. Under physiological conditions, ANG-II role in ROS autophagy-dependent cardiac remodeling in the heart [97].
production and stimulation of redox-mediated signaling Additionally, ANG 1–7 reverses autophagic activation and

13
Pflügers Archiv - European Journal of Physiology

protects the heart from oxidative stress and hypertrophic DCM, we may see a protective mechanism but it requires
response induced by ANG-II [97]. However, the role of detailed investigation.
autophagy in the progression or protection of DCM has
started emerging. It has been shown that during DCM, oxi-
dative stress sensitizes the heart to the activation of RAAS RAAS signaling in obesity and diet
signaling and induces autophagic type-II programmed cell in the development of DCM
death, leading to accelerated cardiac remodeling and dys-
function [7, 168, 188]. Specifically, autophagy in the heart is Lifestyle modifications are one of the self-controlled hab-
induced by type 1 diabetes but inhibited in type 2 diabetes, its to prevent most non-genetic metabolic diseases. Poor
although exact mechanisms are unclear [82]. Independent of diet and physical inactivity are the most influential fac-
this, elevated oxidative stress, inflammation, apoptosis, and tors contributing to an increase in obesity in addition to
ER stress induced by the increased production of monocyte the multiple co-existing conditions such as hypertension,
chemoattractant protein-1 (MCP-1) are suspected for contrib- glucose intolerance, dyslipidemia, and obstructive sleep
uting to the disruption of normal myocardial autophagy in apnea in DCM [83]. Additionally, obesity is associated
DM, which may impair the removal of damaged organelles with LV diastolic dysfunction and increases the liability of
resulting in dysfunctional myocardium [199, 209]. death from cardiovascular disease, including DM, kidney
On the other hand, insulin signaling itself is a disease, and coronary heart diseases [47, 149, 187]. This
regulator of myocardial autophagy [147]. Studies show is supported by the positive clinical outcome of lifestyle
that autophagy protein–deficient mice have diminished modification that has been shown to improve cardiovas-
autophagy and increased the production of ROS, which cular function [187]. Moreover, metabolic and neurohor-
activates NF-κB in infiltrating macrophages, leading to monal mechanisms have also been postulated to contribute
increased cardiac inflammation and cardiac fibrosis [215]. significantly to obesity-induced DCM. It has recently been
Therefore, it can be that normal autophagy signaling shown that a high-fat diet promoted progressive impair-
can protect the heart from hyperglycemia-induced ment of coronary vascular function in Sprague–Dawley
inflammation and cardiac injury [209]. Constitutive rats [89]. In particular, the early stage of obesity is asso-
autophagy is crucial during any development stage, ciated with reduced vascular NO availability [57] and
including the heart, to eliminate nonfunctional or increased production of ROS [156]. However, the patho-
damaged cells. Therefore, a decrease in autophagy results physiological mechanisms linking obesity to coronary vas-
in abnormal protein accumulation, leading to apoptosis cular dysfunction remains poorly understood.
and cardiac dysfunction in DCM [192, 201]. A recent RAAS signaling in adipose tissue has been shown to
study demonstrated that timely removal of damaged play a major role in obesity-induced cardiovascular dis-
mitochondria in cardiomyocytes through autophagy ease [101]. It is known that ANG-II can induce mac-
contributes to adaptive responses that result in net oxidative rophage infiltration into the arterial walls [17, 29], and
stress and inflammation reductions, thus an adaptive macrophages are an essential mediator of adipose tissue
response that maintains homeostasis [128, 161]. As inflammation and contribute to obesity-associated meta-
RAAS contributes to many of the factors such as oxidative bolic dysfunction [129, 194]. Inflammatory cytokines
stress, inflammation, and mitochondrial dysfunction secreted by macrophages may also impair insulin signaling
as described above, its role in regulating autophagy or [94]. The resulting insulin resistance causes hyperglyce-
mitochondrial specific autophagy (mitophagy) in the mia and increased production of AGEs and ROS, which
context of DCM has not been extensively studied, thus results in vasculature damage [38]. As stated previously,
opening a potential area of investigation. However, in chronic ANG 1–7 treatment completely restored diastolic
general, RAAS is shown to be positively associated with function in diabetic db/db mice by suppressing lipotoxic-
autophagy in cardiomyocytes [23, 97]. It has also been ity, adipose inflammation, and insulin resistance [61, 113].
observed that both ANG-II and autophagy are induced Deletion of ACE-2 resulted in a phenotype with increased
during cardiac dysfunction [140]. Altogether, and with epicardial adipose tissue inflammation, lipotoxicity, oxi-
additional evidence, it clearly suggests that activation of dative stress, and cardiac dysfunction [135]. These results
autophagy by RAAS is protective in high-glucose-treated suggest that ANG 1–7 and ACE-2 are negative regulators
endothelial cells [21]. Therefore, activating autophagy of adipose tissue inflammation and cardiac dysfunction in
has been shown to protect cardiomyocytes by decreasing the obese state. Previous studies have also revealed that
ANG-II-induced oxidative stress and inflammation [51]. adiponectin plays an important role in regulating glucose
As RAAS has been shown to promote autophagy in many and lipid metabolism [136], insulin resistance, and vascu-
cardiovascular complications such as ischemia-reperfusion lar injury [50]. Decreased adiponectin levels in the obese
injury and cardiac hypertrophy, we believe that similarly in subjects are associated with insulin resistance, diabetes,

13
Pflügers Archiv - European Journal of Physiology

Fig. 3  Association between
T2DM, ANG-II activation,
and the risk for cardiovascular
diseases. Insulin resistance is
induced in humans through
unhealthy life style, genetic pre-
disposition, medications, envi-
ronmental factors, and aging.
The activation of RAS increases
the oxidative stress and reduces
the NO bioavailability leading
to systemic inflammation and an
increased risk of CVD

and cardiovascular problems [79], where adiponectin oxidative stress by decreasing mitochondrial inflammation
treatment could be beneficial. Adiponectin treatment and DNA damage induced by ROS production [126, 139].
was shown to attenuate ROS-induced inflammation and However, an exogenous supplement of antioxidants is not
autophagy through the downregulation of AMPK/mTOR beneficial in certain conditions, especially during physical
signaling in cardiomyocytes [22, 48]. Also, adiponectin exercise, where it could completely inhibit the activation
reversed endothelial dysfunction by increasing NO produc- of the endogenous self-defense system [148]. Therefore,
tion in obese DM rats [37]. Taken together, in the context understanding how obesity, physical activity, and RAAS
of obesity, RAAS signaling could promote ROS and adi- influence the production of ROS will be critical to manag-
pose tissue inflammation to advance the progression from ing obesity-associated DCM.
obesity to DCM.
Interestingly, there is the interplay between the SNS
and the RAAS in obesity [81, 99], where obesity leads to Perspectives
the activation of the SNS, which activates the production
of renin and angiotensinogen and consequently promotes DCM is recognized as being highly prevalent in the diabetic
excessive ANG-II production. Activation of the SNS in population, and risks for developing the same are higher
obese patients might partially explain the increases in LV compared to cardiomyopathy alone. Hence, early screen-
mass observed in obese patients as an increase in local ing and treatments are necessary to reduce the development
ANG-II production, which could stimulate hypertrophy and progression of DCM, which further reduces the risk of
and fibrosis [2, 4]. Therefore, RAAS and obesity-induced acute myocardial infarction, atherosclerosis, and heart fail-
activation of the SNS in combination with DM undoubtedly ure. Chronic hyperglycemia and insulin resistance lead to
exacerbates DCM. Moreover, a recent study revealed that increased activation of RAAS signaling and vice versa that
the improved outcome from the exercise and diet-induced co-exists with or without oxidative stress, inflammation,
weight loss was through the downregulation of sympathetic autophagy, mitochondrial dysfunction and other co-factors,
nerve activity [169]. Over a 35-year follow-up period in a which induce structural disturbances and functional abnor-
Swedish cohort study, it was shown that long-term physi- malities to the heart (Fig. 3). Experimental evidence support
cal activity positively influences morbidity and mortality the role of ANG 1–7 and antioxidants in reversing many of
in middle-aged participants [18]. Indeed, another study the impairments in the myocardium, suggesting that ANG
revealed that physical activity was independently associ- 1–7 may have a promising approach in the management of
ated with a 26% reduction in the risk of DM [116]. Recent DCM. Given that cardiovascular diseases are the principal
studies also show that in both diabetic and non-diabetic con- cause of morbidity and mortality among DM patients, the
ditions, physical exercise increases endothelium-derived efficacy of new therapies warrants investigation for this
NO production and antioxidant activation and reduces growing patient population.

13
Pflügers Archiv - European Journal of Physiology

Acknowledgements  This study was made possible by a Rapid cardiovascular dysfunction. Am J Physiol Heart Circ Physiol
Response Call (RRC) award (RRC2-076) to H. C. Y. from the 292:H666-672. https://​doi.​org/​10.​1152/​ajphe​art.​00372.​2006
Qatar National Research Fund (a member of The Qatar Founda- 13. Boudina S, Abel ED (2007) Diabetic cardiomyopathy revisited.
tion). Venkatesh Sundararajan’s laboratory work is partially sup- Circulation 115:3213–3223. https://​doi.​org/​10.​1161/​CIRCU​
ported by the American Heart Association grants (20CDA35260096 LATIO​NAHA.​106.​679597 (115/25/3213[pii])
and 20TPA3542000) 14. Boudina S, Abel ED (2010) Diabetic cardiomyopathy, causes
and effects. Rev Endocr Metab Disord 11:31–39. https://​doi.​
org/​10.​1007/​s11154-​010-​9131-7
Declarations  15. Boudina S, Bugger H, Sena S, O’Neill BT, Zaha VG, Ilkun O,
Wright JJ, Mazumder PK, Palfreyman E, Tidwell TJ, Theobald
Conflict of interest  The authors declare no competing interests. H, Khalimonchuk O, Wayment B, Sheng XM, Rodnick KJ,
Centini R, Chen D, Litwin SE, Weimer BE, Abel ED (2009)
Contribution of impaired myocardial insulin signaling to mito-
chondrial dysfunction and oxidative stress in the heart. Cir-
References culation 119:1272-U1111. https://​doi.​org/​10.​1161/​Circu​latio​
naha.​108.​792101
1. Abdellatif M, Sedej S, Carmona-Gutierrez D, Madeo F, Kroemer 16. Bugger H, Riehle C, Jaishy B, Wende AR, Tuinei J, Chen D,
G (2018) Autophagy in cardiovascular aging. Circ Res 123:803– Soto J, Pires KM, Boudina S, Theobald HA, Luptak I, Way-
824. https://​doi.​org/​10.​1161/​CIRCR​ESAHA.​118.​312208 ment B, Wang X, Litwin SE, Weimer BC, Abel ED (2012)
2. Abel ED, Litwin SE, Sweeney G (2008) Cardiac remodeling in Genetic loss of insulin receptors worsens cardiac efficiency in
obesity. Physiol Rev 88:389–419. https://​doi.​org/​10.​1152/​physr​ diabetes. J Mol Cell Cardiol 52:1019–1026. https://​doi.​org/​10.​
ev.​00017.​2007 1016/j.​yjmcc.​2012.​02.​001
3. Acar G, Akcay A, Sokmen A, Ozkaya M, Guler E, Sokmen G, Kaya 17. Bush E, Maeda N, Kuziel WA, Dawson TC, Wilcox JN, DeLeon
H, Nacar AB, Tuncer C (2009) Assessment of atrial electromechan- H, Taylor WR (2000) CC chemokine receptor 2 is required for
ical delay, diastolic functions, and left atrial mechanical functions macrophage infiltration and vascular hypertrophy in angiotensin
in patients with type 1 diabetes mellitus. J Am Soc Echocardiogr II-induced hypertension. Hypertension 36:360–363
22:732–738. https://​doi.​org/​10.​1016/j.​echo.​2009.​03.​028 18. Byberg L, Melhus H, Gedeborg R, Sundstrom J, Ahlbom A, Zethe-
4. Amador N, Encarnacion JD, Rodriguez L, Tello A, Lopez M, lius B, Berglund LG, Wolk A, Michaelsson K (2009) Total mor-
Guizar JM (2004) Relationship between left ventricular mass and tality after changes in leisure time physical activity in 50 year old
heart sympathetic activity in male obese subjects. Arch Med Res men: 35 year follow-up of population based cohort. British Medical
35:411–415. https://​doi.​org/​10.​1016/j.​arcmed.​2004.​05.​003 Journal 338. ARTN b688. https://​doi.​org/​10.​1136/​bmj.​b688
5. Anderson EJ, Kypson AP, Rodriguez E, Anderson CA, Lehr EJ, 19. Candido R, Jandeleit-Dahm KA, Cao Z, Nesteroff SP, Burns WC,
Neufer PD (2009) Substrate-specific derangements in mitochon- Twigg SM, Dilley RJ, Cooper ME, Allen TJ (2002) Prevention
drial metabolism and redox balance in the atrium of the type 2 of accelerated atherosclerosis by angiotensin-converting enzyme
diabetic human heart. J Am Coll Cardiol 54:1891–1898. https://​ inhibition in diabetic apolipoprotein E-deficient mice. Circula-
doi.​org/​10.​1016/j.​jacc.​2009.​07.​031 tion 106:246–253
6. Andreozzi F, Laratta E, Sciacqua A, Perticone F, Sesti G (2004) 20. Castoldi G, di Gioia CRT, Roma F, Carletti R, Manzoni G, Stella
Angiotensin II impairs the insulin signaling pathway promoting A, Zerbini G, Perseghin G (2019) Activation of angiotensin type
production of nitric oxide by inducing phosphorylation of insulin 2 (AT2) receptors prevents myocardial hypertrophy in Zucker
receptor substrate-1 on Ser(312) and Ser(616) in human umbili- diabetic fatty rats. Acta Diabetol 56:97–104. https://​doi.​org/​10.​
cal vein endothelial cells. Circ Res 94:1211–1218. https://​doi.​ 1007/​s00592-​018-​1220-1
org/​10.​1161/​01.​Res.​00001​26501.​34994.​96 21. Chen F, Chen B, Xiao FQ, Wu YT, Wang RH, Sun ZW, Fu
7. Ansley DM, Wang BH (2013) Oxidative stress and myocardial GS, Mou Y, Tao W, Hu XS, Hu SJ (2014) Autophagy protects
injury in the diabetic heart. Journal of Pathology 229:232–241. against senescence and apoptosis via the RAS-mitochondria in
https://​doi.​org/​10.​1002/​path.​4113 high-glucose-induced endothelial cells. Cell Physiol Biochem
8. Atlas SA (2007) The renin-angiotensin aldosterone system: 33:1058–1074. https://​doi.​org/​10.​1159/​00035​8676
pathophysiological role and pharmacologic inhibition. J Manag 22. Cheng J, Wang M, Ma H, Li H, Ren J, Wang R (2015) Adiponec-
Care Pharm 13:9–20. https://​doi.​org/​10.​18553/​jmcp.​2007.​13.​ tin inhibits oxidative stress and modulates TGF-b1 and COL-1
s8-b (2007(13)8:9-20 [pii]) expression via the AMPK pathway in HSC-T6 cells. Zhonghua
9. Banday AA, Lokhandwala MF (2008) Oxidative stress-induced Gan Zang Bing Za Zhi 23:69–72
renal angiotensin AT1 receptor upregulation causes increased 23. Cheng Z, Zhang M, Hu J, Lin J, Feng X, Wang S, Wang T, Gao
stimulation of sodium transporters and hypertension. Am J Phys- E, Wang H, Sun D (2018) Mst1 knockout enhances cardiomyo-
iol Renal Physiol 295:F698-706. https://d​ oi.o​ rg/1​ 0.1​ 152/a​ jpren​ al.​ cyte autophagic flux to alleviate angiotensin II-induced cardiac
90308.​2008 (90308.2008[pii]) injury independent of angiotensin II receptors. J Mol Cell Cardiol
10. Banday AA, Lokhandwala MF (2011) Oxidative stress causes 125:117–128. https://​doi.​org/​10.​1016/j.​yjmcc.​2018.​08.​028
renal angiotensin II type 1 receptor upregulation, Na+/H+ 24. Chow BSM, Koulis C, Krishnaswamy P, Steckelings UM, Unger
exchanger 3 overstimulation, and hypertension. Hypertension T, Cooper ME, Jandeleit-Dahm KA, Allen TJ (2016) The angio-
57:452–459. https://​doi.​org/​10.​1161/​HYPER​TENSI​ONAHA.​ tensin II type 2 receptor agonist Compound 21 is protective in
110.​162339 (HYPERTENSIONAHA.110.162339[pii]) experimental diabetes-associated atherosclerosis. Diabetologia
11. Benigni A, Corna D, Zoja C, Sonzogni A, Latini R, Salio M, 59:1778–1790. https://​doi.​org/​10.​1007/​s00125-​016-​3977-5
Conti S, Rottoli D, Longaretti L, Cassis P, Morigi M, Coffman 25. Connelly KA, Kelly DJ, Zhang Y, Prior DL, Martin J, Cox AJ,
TM, Remuzzi G (2009) Disruption of the Ang II type 1 recep- Thai K, Feneley MP, Tsoporis J, White KE, Krum H, Gilbert
tor promotes longevity in mice. J Clin Investig 119:524–530. RE (2007) Functional, structural and molecular aspects of dias-
https://​doi.​org/​10.​1172/​JCI36​703 tolic heart failure in the diabetic (mRen-2)27 rat. Cardiovasc
12. Benter IF, Yousif MH, Cojocel C, Al-Maghrebi M, Diz Res 76:280–291. https://​doi.​org/​10.​1016/j.​cardi​ores.​2007.​06.​
DI (2007) Angiotensin-(1–7) prevents diabetes-induced 022

13
Pflügers Archiv - European Journal of Physiology

26. Conti V, Izzo V, Corbi G, Russomanno G, Manzo V, De Lise F, 41. Dinh W, Futh R, Nickl W, Krahn T, Ellinghaus P, Scheffold T,
Di Donato A, Filippelli A (2016) Antioxidant supplementation Bansemir L, Bufe A, Barroso MC, Lankisch M (2009) Elevated
in the treatment of aging-associated diseases. Frontiers in Phar- plasma levels of TNF-alpha and Interleukin-6 in patients with
macology 7. Artn 24. https://​doi.​org/​10.​3389/​Fphar.​2016.​00024 diastolic dysfunction and glucose metabolism disorders. Car-
27. Crabos M, Roth M, Hahn AWA, Erne P (1994) Characterization diovascular Diabetology 8. Artn 58. https://​doi.​org/​10.​1186/​
of angiotensin-ii receptors in cultured adult-rat cardiac fibro- 1475-​2840-8-​58
blasts - coupling to signaling systems and gene-expression. J 42. Donath MY, Storling J, Maedler K, Mandrup-Poulsen T (2003)
Clin Investig 93:2372–2378. https://​doi.​org/​10.​1172/​Jci11​7243 Inflammatory mediators and islet beta-cell failure: a link between
28. Crowley SD, Coffman TM (2012) Recent advances involving the type 1 and type 2 diabetes. J Mol Med (Berl) 81:455–470. https://​
renin-angiotensin system. Exp Cell Res 318:1049–1056. https://​ doi.​org/​10.​1007/​s00109-​003-​0450-y
doi.​org/​10.​1016/j.​yexcr.​2012.​02.​023 43. Doughan AK, Harrison DG, Dikalov SI (2008) Molecular mech-
29. Dai Q, Xu M, Yao M, Sun B (2007) Angiotensin AT1 recep- anisms of angiotensin II-mediated mitochondrial dysfunction:
tor antagonists exert antiinflammatory effects in spontaneously linking mitochondrial oxidative damage and vascular endothe-
hypertensive rats. Br J Pharmacol 152:1042–1048. https://​doi.​ lial dysfunction. Circ Res 102:488–496. https://​doi.​org/​10.​1161/​
org/​10.​1038/​sj.​bjp.​07074​54 CIRCR​ESAHA.​107.​162800
30. Daly CA, Fox KM, Remme WJ, Bertrand ME, Ferrari R, 44. Durand MJ, Zinkevich NS, Riedel M, Gutterman DD, Nasci VL,
Simoons ML (2005) The effect of perindopril on cardiovas- Salato VK, Hijjawi JB, Reuben CF, North PE, Beyer AM (2016)
cular morbidity and mortality in patients with diabetes in the Vascular actions of angiotensin 1–7 in the human microcircula-
EUROPA study: results from the PERSUADE substudy. Eur tion novel role for telomerase. Arterioscler Thromb Vasc Biol
Heart J 26:1369–1378. https://​doi.​org/​10.​1093/​eurhe​artj/​ehi225 36:1254-+. https://​doi.​org/​10.​1161/​Atvba​ha.​116.​307518
(ehi225[pii]) 45. Dzau V (2005) The cardiovascular continuum and renin-angi-
31. Dandamudi S, Slusser J, Mahoney DW, Redfield MM, Rodeheffer otensin-aldosterone system blockade. J Hypertens 23:S9–S17.
RJ, Chen HH (2014) The prevalence of diabetic cardiomyopathy: https://​doi.​org/​10.​1097/​01.​hjh.​00001​65623.​72310.​dd
a population-based study in Olmsted County, Minnesota. J Car- 46. Ebermann L, Spillmann F, Sidiropoulos M, Escher F, Heringer-
diac Fail 20:304–309. https://​doi.​org/​10.​1016/j.​cardf​ail.​2014.​02.​ Walther S, Schultheiss HP, Tschope C, Walther T (2008) The
007 angiotensin-(1–7) receptor agonist AVE0991 is cardioprotective
32. de Gasparo M, Catt KJ, Inagami T, Wright JW, Unger T (2000) in diabetic rats. Eur J Pharmacol 590:276–280. https://​doi.​org/​
International union of pharmacology. XXIII. The angiotensin II 10.​1016/j.​ejphar.​2008.​05.​024
receptors. Pharmacol Rev 52:415–472 47. Eckel RH (2008) Nonsurgical management of obesity in adults.
33. De Mello WC (2004) Angiotensin (1–7) re-establishes impulse N Engl J Med 358:1941–1950. https://​doi.​org/​10.​1056/​Nejmc​
conduction in cardiac muscle during ischaemia-reperfusion. The p0801​652
role of the sodium pump. J Renin-Angiotensin-Aldosterone Syst 48. Essick EE, Wilson RM, Pimentel DR, Shimano M, Baid S, Ouchi
5:203–208. https://​doi.​org/​10.​3317/​jraas.​2004.​041 N, Sam F (2013) Adiponectin modulates oxidative stress-induced
34. De Mello WC (2014) Angiotensin (1–7) re-establishes heart cell autophagy in cardiomyocytes. Plos One 8. UNSP e68697. https://​
communication previously impaired by cell swelling: Implica- doi.​org/​10.​1371/​journ​al.​pone.​00686​97
tions for myocardial ischemia. Exp Cell Res 323:359–365. 49. Fang ZY, Prins JB, Marwick TH (2004) Diabetic cardiomyopa-
https://​doi.​org/​10.​1016/j.​yexcr.​2014.​03.​006 thy: evidence, mechanisms, and therapeutic implications. Endocr
35. De Mello WC, Danser AHJ (2000) Angiotensin II and the heart Rev 25:543–567. https://​doi.​org/​10.​1210/​er.​2003-​0012.​25/4/​
- on the intracrine renin-angiotensin system. Hypertension 543[pii]
35:1183–1188 50. Fenger M (2013) The enigma of adiponectin. Atherosclerosis
36. De Mello WC, Gerena Y (2010) Further studies on the effects 227:226–227. https://​doi.​org/​10.​1016/j.​ather​oscle​rosis.​2012.​11.​018
of intracrine and extracellular angiotensin II on the regulation of 51. Finckenberg P, Eriksson O, Baumann M, Merasto S, Lalowski
heart cell volume. On the influence of aldosterone and spirono- MM, Levijoki J, Haasio K, Kyto V, Muller DN, Luft FC, Oresic
lactone. Regul Pept 165:200–205. https://​doi.​org/​10.​1016/j.​reg- M, Mervaala E (2012) Caloric restriction ameliorates angiotensin
pep.​2010.​07.​165 II-induced mitochondrial remodeling and cardiac hypertrophy.
37. Deng G, Long Y, Yu YR, Li MR (2010) Adiponectin directly Hypertension 59:76–84. https://​doi.​org/​10.​1161/​HYPER​TENSI​
improves endothelial dysfunction in obese rats through the ONAHA.​111.​179457
AMPK-eNOS Pathway. Int J Obes 34:165–171. https://​doi.​org/​ 52. Fiordaliso F, Cuccovillo I, Bianchi R, Bai A, Doni M, Salio M,
10.​1038/​ijo.​2009.​205 De Angelis N, Ghezzi P, Latini R, Masson S (2006) Cardiovascu-
38. Di Marco E, Gray SP, Jandeleit-Dahm K (2013) Diabetes alters lar oxidative stress is reduced by an ACE inhibitor in a rat model
activation and repression of pro- and anti-inflammatory signaling of streptozotocin-induced diabetes. Life Sci 79:121–129. https://​
pathways in the vasculature. Front Endocrinol (Lausanne) 4:68. doi.​org/​10.​1016/j.​lfs.​2005.​12.​036
https://​doi.​org/​10.​3389/​fendo.​2013.​00068 53. Flather MD, Yusuf S, Kober L, Pfeffer M, Hall A, Murray G,
39. Diamant M, Lamb HJ, Groeneveld Y, Endert EL, Smit JWA, Torp-Pedersen C, Ball S, Pogue J, Moye L, Braunwald E, C
Bax JJ, Romijn JA, de Roos A, Radder JK (2003) Diastolic dys- AIMI, (2000) Long-term ACE-inhibitor therapy in patients with
function is associated with altered myocardial metabolism in heart failure or left-ventricular dysfunction: a systematic over-
asymptomatic normotensive patients with well-controlled type view of data from individual patients. Lancet 355:1575–1581.
2 diabetes mellitus. J Am Coll Cardiol 42:328–335. https://​doi.​ https://​doi.​org/​10.​1016/​S0140-​6736(00)​02212-1
org/​10.​1016/​S0735-​1097(03)​00625-9 54. Frustaci A, Kajstura J, Chimenti C, Jakoniuk I, Leri A, Maseri
40. Dias-Peixoto MF, Santos RAS, Gomes ERM, Alves MNM, A, Nadal-Ginard B, Anversa P (2000) Myocardial cell death in
Almeida PWM, Greco L, Rosa M, Fauler B, Bader M, Alenina human diabetes. Circ Res 87:1123–1132
N, Guatimosim S (2008) Molecular mechanisms involved in the 55. Fukuda T, Ahearn M, Roberts A, Mattaliano RJ, Zaal K, Ralston
angiotensin-(1–7)/Mas signaling pathway in cardiomyocytes. E, Plotz PH, Raben N (2006) Autophagy and mistargeting of
Hypertension 52:542–548. https://​doi.​org/​10.​1161/​Hyper ​tensi​ therapeutic enzyme in skeletal muscle in Pompe disease. Mol
onaha.​108.​114280 Ther 14:831–839. https://​doi.​org/​10.​1016/j.​ymthe.​2006.​08.​009

13
Pflügers Archiv - European Journal of Physiology

56. Galderisi M, Anderson KM, Wilson PW, Levy D (1991) Echo- 70. Heart Outcomes Prevention Evaluation Study Investigators
cardiographic evidence for the existence of a distinct diabetic (2000) Effects of ramipril on cardiovascular and microvascular
cardiomyopathy (the Framingham Heart Study). Am J Cardiol outcomes in people with diabetes mellitus: results of the HOPE
68:85–89 (0002-9149(91)90716-X[pii]) study and MICRO-HOPE substudy. Lancet 355(9200):253–9.
57. Gamez-Mendez AM, Vargas-Robles H, Rios A, Escalante B https://​doi.​org/​10.​1016/​S0140-​6736(99)​12323-7
(2015) Oxidative stress-dependent coronary endothelial dysfunc- 71. Hussein SA, El-Wadood HM, Abdallah MA, Khorshed AA
tion in obese mice. Plos One 10. ARTN e0138609. https://​doi.​ (2015) HPTLC with fluorescence densitometry for simultane-
org/​10.​1371/​journ​al.​pone.​01386​09 ous determination of some angiotensin II receptor blockers in
58. Gao XM, Tsai A, Al-Sharea A, Su Y, Moore S, Han LP, Kiriazis tablets and plasma. J AOAC Int 98:354–360. https://​doi.​org/​
H, Dart AM, Murphy AJ, Du XJ (2017) Inhibition of the renin- 10.​5740/​jaoac​int.​14-​179
angiotensin system post myocardial infarction prevents inflam- 72. Huynh K, Kiriazis H, Du XJ, Love JE, Jandeleit-Dahm KA,
mation-associated acute cardiac rupture. Cardiovasc Drugs Ther Forbes JM, McMullen JR, Ritchie RH (2012) Coenzyme Q10
31:145–156. https://​doi.​org/​10.​1007/​s10557-​017-​6717-​2[pii] attenuates diastolic dysfunction, cardiomyocyte hypertrophy
59. Gao YL, Kang L, Li CM, Wang XY, Sun CF, Li QZ, Liu RH, and cardiac fibrosis in the db/db mouse model of type 2 dia-
Wang JP (2016) Resveratrol ameliorates diabetes-induced car- betes. Diabetologia 55:1544–1553. https://​d oi.​o rg/​1 0.​1 007/​
diac dysfunction through AT1R-ERK/p38 MAPK signaling path- s00125-​012-​2495-3
way. Cardiovasc Toxicol 16:130–137. https://​doi.​org/​10.​1007/​ 73. Ibsen H, Lindholm LH, Pedersen OL, Dahlof B, Kjeldsen S
s12012-​015-​9321-3 (2003) The effect of losartan versus atenolol on cardiovascular
60. Giani JF, Gironacci MM, Munoz MC, Pena C, Turyn D, morbidity and mortality in patients with diabetes mellitus in
Dominici FP (2007) Angiotensin-(1 7) stimulates the phospho- the LIFE-study. Ugeskr Laeger 165:459–462
rylation of JAK2, IRS-1 and Akt in rat heart in vivo: role of 74. Jager J, Gremeaux T, Cormont M, Le Marchand-Brustel Y,
the AT1 and Mas receptors. Am J Physiol Heart Circ Physiol Tanti JF (2007) Interleukin-1beta-induced insulin resistance
293:H1154-1163. https://​doi.​org/​10.​1152/​ajphe​art.​01395.​2006 in adipocytes through down-regulation of insulin receptor sub-
(01395.2006[pii]) strate-1 expression. Endocrinology 148:241–251. https://​doi.​
61. Giani JF, Mayer MA, Munoz MC, Silberman EA, Hocht C, Taira org/​10.​1210/​en.​2006-​0692 (en.2006-0692[pii])
CA, Gironacci MM, Turyn D, Dominici FP (2009) Chronic infu- 75. Jia G, DeMarco VG, Sowers JR (2016) Insulin resistance and
sion of angiotensin-(1–7) improves insulin resistance and hyper- hyperinsulinaemia in diabetic cardiomyopathy. Nat Rev Endo-
tension induced by a high-fructose diet in rats. Am J Physiol crinol 12:144–153. https://​doi.​org/​10.​1038/​nrendo.​2015.​216
Endocrinol Metab 296:E262-271. https://​doi.​org/​10.​1152/​ajpen​ 76. Jia G, Whaley-Connell A, Sowers JR (2018) Diabetic cardio-
do.​90678.​2008 (90678.2008[pii]) myopathy: a hyperglycaemia- and insulin-resistance-induced
62. Goldin A, Beckman JA, Schmidt AM, Creager MA (2006) heart disease. Diabetologia 61:21–28. https://​doi.​org/​10.​1007/​
Advanced glycation end products - sparking the development of s00125-​017-​4390-4
diabetic vascular injury. Circulation 114:597–605. https://​doi.​ 77. Jiang F, Yang J, Zhang Y, Dong M, Wang S, Zhang Q, Liu FF,
org/​10.​1161/​Circu​latio​naha.​106.​621854 Zhang K, Zhang C (2014) Angiotensin-converting enzyme 2
63. Grobe JL, Mecca AP, Lingis M, Shenoy V, Bolton TA, Machado and angiotensin 1–7: novel therapeutic targets. Nat Rev Car-
JM, Speth RC, Raizada MK, Katovich MJ (2007) Prevention of diol 11:413–426. https://​d oi.​o rg/​1 0.​1 038/​n rcar​d io.​2 014.​5 9
angiotensin II-induced cardiac remodeling by angiotensin-(1–7). (nrcardio.2014.59[pii])
Am J Physiol Heart Circ Physiol 292:H736-742. https://​doi.​org/​ 78. Kadoguchi T, Kinugawa S, Takada S, Fukushima A, Furihata T,
10.​1152/​ajphe​art.​00937.​2006 Homma T, Masaki Y, Mizushima W, Nishikawa M, Takahashi
64. Guariguata L, Whiting DR, Hambleton I, Beagley J, Linnen- M, Yokota T, Matsushima S, Okita K, Tsutsui H (2015) Angio-
kamp U, Shaw JE (2014) Global estimates of diabetes preva- tensin II can directly induce mitochondrial dysfunction, decrease
lence for 2013 and projections for 2035. Diabetes Res Clin Pract oxidative fibre number and induce atrophy in mouse hindlimb
103:137–149. https://​doi.​org/​10.​1016/j.​diabr​es.​2013.​11.​002 skeletal muscle. Exp Physiol 100:312–322. https://​doi.​org/​10.​
(S0168-8227(13)00385-9[pii]) 1113/​expph​ysiol.​2014.​084095
65. Ha H, Hwang IA, Park JH, Lee HB (2008) Role of reactive oxygen 79. Kadowaki T, Yamauchi T, Kubota N, Hara K, Ueki K, Tobe K
species in the pathogenesis of diabetic nephropathy. Diabetes Res (2006) Adiponectin and adiponectin receptors in insulin resist-
Clin Pract 82:S42–S45. https://​doi.​org/​10.​1016/j.​diabr​es.​2008.​09.​017 ance, diabetes, and the metabolic syndrome. J Clin Investig
66. Ha JW, Lee HC, Kang ES, Ahn CM, Kim JM, Ahn JA, Lee 116:1784–1792. https://​doi.​org/​10.​1172/​JCI29​126
SW, Choi EY, Rim SJ, Oh JK, Chung N (2007) Abnormal left 80. Kajstura J, Fiordaliso F, Andreoli AM, Li B, Chimenti S, Medow
ventricular longitudinal functional reserve in patients with dia- MS, Limana F, Nadal-Ginard B, Leri A, Anversa P (2001) IGF-1
betes mellitus: implication for detecting subclinical myocardial overexpression inhibits the development of diabetic cardiomyo-
dysfunction using exercise tissue Doppler echocardiography. pathy and angiotensin II-mediated oxidative stress. Diabetes
Heart 93:1571–1576. https://​doi.​org/​10.​1136/​hrt.​2006.​101667 50:1414–1424. https://​doi.​org/​10.​2337/​diabe​tes.​50.6.​1414
(hrt.2006.101667[pii]) 81. Kalil GZ, Haynes WG (2012) Sympathetic nervous system in
67. Haffner SM (2006) The metabolic syndrome: inflammation, obesity-related hypertension: mechanisms and clinical implica-
diabetes mellitus, and cardiovascular disease. Am J Cardiol tions. Hypertens Res 35:4–16. https://​doi.​org/​10.​1038/​hr.​2011.​
97:3A-11A. https://​d oi.​o rg/​1 0.​1 016/j.​a mjca​r d.​2 005.​1 1.​0 10 173
(S0002-9149(05)02028-X[pii]) 82. Kanamori H, Takemura G, Goto K, Tsujimoto A, Mikami A,
68. Hao PP, Yang JM, Zhang MX, Zhang K, Chen YG, Zhang C, Ogino A, Watanabe T, Morishita K, Okada H, Kawasaki M,
Zhang Y (2015) Angiotensin-(1–7) treatment mitigates right Seishima M, Minatoguchi S (2015) Autophagic adaptations in
ventricular fibrosis as a distinctive feature of diabetic cardio- diabetic cardiomyopathy differ between type 1 and type 2 dia-
myopathy. Am J Physiol Heart Circ Physiol 308:H1007-1019. betes. Autophagy 11:1146–1160. https://​doi.​org/​10.​1080/​15548​
https://​doi.​org/​10.​1152/​ajphe​art.​00563.​2014 627.​2015.​10512​95
69. Hayat SA, Patel B, Khattar RS, Malik RA (2004) Diabetic 83. Karam JG, McFarlane SI (2011) Update on the prevention of
cardiomyopathy: mechanisms, diagnosis and treatment. Clin type 2 diabetes. Curr DiabRep 11:56–63. https://d​ oi.o​ rg/1​ 0.1​ 007/​
Sci 107:539–557 s11892-​010-​0163-x

13
Pflügers Archiv - European Journal of Physiology

84. Kassiri Z, Zhong JC, Guo D, Basu R, Wang XH, Liu PP, Scholey 98. Lindholm LH, Ibsen H, Dahlof B, Devereux RB, Beevers G, de
JW, Penninger JM, Oudit GY (2009) Loss of angiotensin-convert- Faire U, Fyhrquist F, Julius S, Kjeldsen SE, Kristiansson K, Led-
ing enzyme 2 accelerates maladaptive left ventricular remodeling erballe-Pedersen O, Nieminen MS, Omvik P, Oparil S, Wedel H,
in response to myocardial infarction. Circulation-Heart Failure Aurup P, Edelman J, Snapinn S, grp Ls, (2002) Cardiovascular
2:446–455. https://​doi.​org/​10.​1161/​Circh​eartf​ailure.​108.​840124 morbidity and mortality in patients with diabetes in the Losar-
85. Kayama Y, Raaz U, Jagger A, Adam M, Schellinger IN, Saka- tan Intervention For Endpoint reduction in hypertension study
moto M, Suzuki H, Toyama K, Spin JM, Tsao PS (2015) Diabetic (LIFE): a randomised trial against atenolol. Lancet 359:1004–
cardiovascular disease induced by oxidative stress. Int J Mol Sci 1010. https://​doi.​org/​10.​1016/​S0140-​6736(02)​08090-X
16:25234–25263. https://​doi.​org/​10.​3390/​ijms1​61025​234 99. Lohmeier TE, Iliescu R (2013) The sympathetic nervous system
86. Khullar M, Al-Shudiefat AA, Ludke A, Binepal G, Singal PK in obesity hypertension. Curr Hypertens Rep 15:409–416. https://​
(2010) Oxidative stress: a key contributor to diabetic cardiomyo- doi.​org/​10.​1007/​s11906-​013-​0356-1
pathy. Can J Physiol Pharmacol 88:233–240. https://​doi.​org/​10.​ 100. Low Wang CC, Hess CN, Hiatt WR, Goldfine AB (2016) Clinical
1139/​Y10-​016 (y10-016[pii]) update: cardiovascular disease in diabetes mellitus: atheroscle-
87. Kim S, Iwao H (2000) Molecular and cellular mechanisms of rotic cardiovascular disease and heart failure in type 2 diabetes
angiotensin II-mediated cardiovascular and renal diseases. Phar- mellitus - mechanisms, management, and clinical considerations.
macol Rev 52:11–34 Circulation 133:2459–2502. https://​doi.​org/​10.​1161/​CIRCU​
88. Kimura S, Zhang GX, Nishiyama A, Shokoji T, Yao L, Fan YY, LATIO​NAHA.​116.​022194 (CIRCULATIONAHA.116.02219
Rahman M, Abe Y (2005) Mitochondria-derived reactive oxygen 4[pii])
species and vascular MAP kinases: comparison of angiotensin 101. Luong KVQ, Nguyen LTH (2013) The beneficial role of vita-
II and diazoxide. Hypertension 45:438–444. https://​doi.​org/​10.​ min D in obesity: possible genetic and cell signaling mecha-
1161/​01.​HYP.​00001​57169.​27818.​ae nisms. Nutrition Journal 12. Artn 89. https://​doi.​org/​10.​1186/​
89. Kleinschmidt TL, Oltman CL (2014) Progression and reversal 1475-​2891-​12-​89
of coronary and mesenteric vascular dysfunction associated with 102. Ma H, Li SY, Xu PS, Babcock SA, Dolence EK, Brownlee M,
obesity. Obesity 22:2193–2200. https://​doi.​org/​10.​1002/​oby.​ Li J, Ren J (2009) Advanced glycation endproduct (AGE) accu-
20837 mulation and AGE receptor (RAGE) up-regulation contribute to
90. Kobori H, Mori H, Masaki T, Nishiyama A (2013) Angiotensin the onset of diabetic cardiomyopathy. J Cell Mol Med 13:1751–
II blockade and renal protection. Curr Pharm Des 19:3033–3042 1764. https://​doi.​org/​10.​1111/j.​1582-​4934.​2008.​00547.x
91. Kritchevsky SB, Cesari M, Pahor M (2005) Inflammatory mark- 103. Ma TK, Kam KK, Yan BP, Lam YY (2010) Renin-angiotensin-
ers and cardiovascular health in older adults. Cardiovasc Res aldosterone system blockade for cardiovascular diseases: current
66:265–275. https://​doi.​org/​10.​1016/j.​cardi​ores.​2004.​12.​026 status. Br J Pharmacol 160:1273–1292. https://d​ oi.o​ rg/1​ 0.1​ 111/j.​
92. Lakshmanan AP, Harima M, Sukumaran V, Soetikno V, Thanda- 1476-​5381.​2010.​00750.x (BPH750[pii])
varayan RA, Suzuki K, Kodama M, Nagata M, Takagi R, Wata- 104. Maalouf RM, Eid AA, Gorin YC, Block K, Escobar GP, Bailey S,
nabe K (2012) Modulation of AT-1R/AMPK-MAPK cascade Abboud HE (2012) Nox4-derived reactive oxygen species medi-
plays crucial role for the pathogenesis of diabetic cardiomyo- ate cardiomyocyte injury in early type 1 diabetes. Am J Physiol
pathy in transgenic type 2 diabetic (Spontaneous Diabetic Toni) Cell Physiol 302:C597–C604. https://​doi.​org/​10.​1152/​ajpce​ll.​
rats. Biochem Pharmacol 83:653–660. https://​doi.​org/​10.​1016/j.​ 00331.​2011
bcp.​2011.​11.​018 105. Makino A, Dai A, Han Y, Youssef KD, Wang W, Dontham-
93. Lautner RQ, Villela DC, Fraga-Silva RA, Silva N, Verano- setty R, Scott BT, Wang H, Dillmann WH (2015) O-GlcNAcase
Braga T, Costa-Fraga F, Jankowski J, Jankowski V, Sousa F, overexpression reverses coronary endothelial cell dysfunction in
Alzamora A, Soares E, Barbosa C, Kjeldsen F, Oliveira A, type 1 diabetic mice. Am J Physiol Cell Physiol 309:C593-599.
Braga J, Savergnini S, Maia G, Peluso AB, Passos-Silva D, Fer- https://​doi.​org/​10.​1152/​ajpce​ll.​00069.​2015
reira A, Alves F, Martins A, Raizada M, Paula R, Motta-Santos 106. Mapanga RF, Essop MF (2016) Damaging effects of hyperglyce-
D, Kemplin F, Pimenta A, Alenina N, Sinisterra R, Bader M, mia on cardiovascular function: spotlight on glucose metabolic
Campagnole-Santos MJ, Santos RAS (2013) Discovery and pathways. American Journal of Physiology-Heart and Circula-
characterization of alamandine a novel component of the renin- tory Physiology 310:H153–H173. https://​doi.​org/​10.​1152/​ajphe​
angiotensin system. Circ Res 112:1104-+. https://​doi.​org/​10.​ art.​00206.​2015
1161/​Circr​esaha.​113.​301077 107. Mariappan N, Elks CM, Sriramula S, Guggilam A, Liu Z, Borkh-
94. Lee J (2013) Adipose tissue macrophages in the development senious O, Francis J (2010) NF-kappaB-induced oxidative stress
of obesity-induced inflammation, insulin resistance and type 2 contributes to mitochondrial and cardiac dysfunction in type II
Diabetes. Arch Pharmacal Res 36:208–222. https://​doi.​org/​10.​ diabetes. Cardiovasc Res 85:473–483. https://​doi.​org/​10.​1093/​
1007/​s12272-​013-​0023-8 cvr/​cvp305 (cvp305[pii])
95. Lei SQ, Li YA, Liu HM, Yu H, Wang H, Xia ZY (2012) Effects 108. Martin-Timon I, Sevillano-Collantes C, Segura-Galindo A, Del
of N-acetylcysteine on nicotinamide dinucleotide phosphate oxi- Canizo-Gomez FJ (2014) Type 2 diabetes and cardiovascular
dase activation and antioxidant status in heart, lung, liver and disease: have all risk factors the same strength? World J Diabetes
kidney in streptozotocin-induced diabetic rats. Yonsei Med J 5:444–470. https://​doi.​org/​10.​4239/​wjd.​v5.​i4.​444
53:294–303. https://​doi.​org/​10.​3349/​ymj.​2012.​53.2.​294 109. Matsusaka H, Kinugawa S, Ide T, Matsushima S, Shiomi T,
96. Lim HS, MacFadyen RJ, Lip GY (2004) Diabetes mellitus, the Kubota T, Sunagawa K, Tsutsui H (2006) Angiotensin II type 1
renin-angiotensin-aldosterone system, and the heart. Arch Intern receptor blocker attenuates exacerbated left ventricular remod-
Med 164:1737–1748. https://​doi.​org/​10.​1001/​archi​nte.​164.​16.​ eling and failure in diabetes-associated myocardial infarction. J
1737.​164/​16/​1737[pii] Cardiovasc Pharmacol 48:95–102. https://d​ oi.o​ rg/1​ 0.1​ 097/0​ 1.fj
​ c.​
97. Lin L, Liu X, Xu J, Weng L, Ren J, Ge J, Zou Y (2016) Mas 00002​45405.​41317.​60
receptor mediates cardioprotection of angiotensin-(1–7) against 110. Mehta PK, Griendling KK (2007) Angiotensin II cell signaling:
angiotensin II-induced cardiomyocyte autophagy and cardiac physiological and pathological effects in the cardiovascular sys-
remodelling through inhibition of oxidative stress. J Cell Mol tem. Am J Physiol Cell Physiol 292:C82–C97. https://d​ oi.o​ rg/1​ 0.​
Med 20:48–57. https://​doi.​org/​10.​1111/​jcmm.​12687 1152/​ajpce​ll.​00287.​2006

13
Pflügers Archiv - European Journal of Physiology

111. Miki T, Yuda S, Kouzu H, Miura T (2013) Diabetic cardiomyo- diabetic cardiovascular disease. Res Cardiovasc Med 4:e26949.
pathy: pathophysiology and clinical features. Heart Fail Rev https://​doi.​org/​10.​5812/​cardi​ovasc​med.​4(2)​2015.​26949
18:149–166. https://​doi.​org/​10.​1007/​s10741-​012-​9313-3 124. Neri Serneri GG, Boddi M, Modesti PA, Coppo M, Cecioni
112. Montezano AC, Touyz RM (2014) Reactive oxygen species, vas- I, Toscano T, Papa ML, Bandinelli M, Lisi GF, Chiavarelli
cular noxs, and hypertension: focus on translational and clinical M (2004) Cardiac angiotensin II participates in coronary
research. Antioxid Redox Signal 20:164–182. https://​doi.​org/​10.​ microvessel inflammation of unstable angina and strengthens
1089/​ars.​2013.​5302 the immunomediated component. Circ Res 94:1630–1637.
113. Mori J, Patel VB, Alrob OA, Basu R, Altamimi T, DesAulniers https://​d oi.​o rg/​1 0.​1 161/​0 1.​R ES.​0 0001​3 0944.​4 9657.​b 8 (01.
J, Wagg CS, Kassiri Z, Lopaschuk GD, Oudit GY (2014) Angi- RES.0000130944.49657.b8[pii])
otensin 1–7 ameliorates diabetic cardiomyopathy and diastolic 125. Nishino I, Fu J, Tanji K, Yamada T, Shimojo S, Koori T, Mora
dysfunction in db/db mice by reducing lipotoxicity and inflam- M, Riggs JE, Oh SJ, Koga Y, Sue CM, Yamamoto A, Murakami
mation. Circulation-Heart Failure 7:327–339. https://​doi.​org/​10.​ N, Shanske S, Byrne E, Bonilla E, Nonaka I, DiMauro S, Hirano
1161/​Circh​eartf​ailure.​113.​000672 M (2000) Primary LAMP-2 deficiency causes X-linked vacu-
114. Morino K, Petersen KF, Dufour S, Befroy D, Frattini J, Shatzkes olar cardiomyopathy and myopathy (Danon disease). Nature
N, Neschen S, White MF, Bilz S, Sono S, Pypaert M, Shulman 406:906–910. https://​doi.​org/​10.​1038/​35022​604
GI (2005) Reduced mitochondrial density and increased IRS-1 126. Nosarev AV, Smagliy LV, Anfinogenova Y, Popov SV, Kapilevich
serine phosphorylation in muscle of insulin-resistant offspring LV (2014) Exercise and NO production: relevance and implica-
of type 2 diabetic parents. J Clin Invest 115:3587–3593. https://​ tions in the cardiopulmonary system. Front Cell Dev Biol 2:73.
doi.​org/​10.​1172/​JCI25​151 https://​doi.​org/​10.​3389/​fcell.​2014.​00073
115. Mozaffarian D, Benjamin EJ, Go AS, Arnett DK, Blaha MJ, 127. Oak JH, Cai H (2007) Attenuation of angiotensin II signaling
Cushman M, Das SR, de Ferranti S, Despres JP, Fullerton HJ, recouples eNOS and inhibits nonendothelial NOX activity in
Howard VJ, Huffman MD, Isasi CR, Jimenez MC, Judd SE, diabetic mice. Diabetes 56:118–126. https://​doi.​org/​10.​2337/​
Kissela BM, Lichtman JH, Lisabeth LD, Liu S, Mackey RH, db06-​0288 (56/1/118[pii])
Magid DJ, McGuire DK, Mohler ER 3rd, Moy CS, Muntner P, 128. Oka T, Hikoso S, Yamaguchi O, Taneike M, Takeda T, Tamai
Mussolino ME, Nasir K, Neumar RW, Nichol G, Palaniappan T, Oyabu J, Murakawa T, Nakayama H, Nishida K, Akira S,
L, Pandey DK, Reeves MJ, Rodriguez CJ, Rosamond W, Sorlie Yamamoto A, Komuro I, Otsu K (2012) Mitochondrial DNA that
PD, Stein J, Towfighi A, Turan TN, Virani SS, Woo D, Yeh RW, escapes from autophagy causes inflammation and heart failure.
Turner MB (2016) Heart Disease And Stroke Statistics-2016 Nature 485:251-U142. https://​doi.​org/​10.​1038/​natur​e10992
update: a report from the American Heart Association. Circu- 129. Olefsky JM, Glass CK (2010) Macrophages, inflammation, and
lation 133:e38-360. https://​doi.​org/​10.​1161/​CIR.​00000​00000​ insulin resistance. Annu Rev Physiol 72:219–246. https://d​ oi.o​ rg/​
000350 (CIR.0000000000000350[pii]) 10.​1146/​annur​ev-​physi​ol-​021909-​135846
116. Mozaffarian D, Kamineni A, Carnethon M, Djousse L, Muka- 130. Oudit GY, Kassiri Z, Patel MP, Chappell M, Butany J, Backx
mal KJ, Siscovick D (2009) Lifestyle risk factors and new-onset PH, Tsushima RG, Scholey JW, Khokha R, Penninger JM (2007)
diabetes mellitus in older adults the cardiovascular health study. Angiotensin II-mediated oxidative stress and inflammation medi-
Arch Intern Med 169:798–807 ate the age-dependent cardiomyopathy in ACE2 null mice. Car-
117. Muhammad Z, Hashmi A (2013) Frequency of diabetic cardio- diovasc Res 75:29–39. https://​doi.​org/​10.​1016/j.​cardi​ores.​2007.​
myopathy among type-2 diabetics presenting as heart failure. 04.​007
Jcpsp-J Coll Physicians Surg Pakistan 23:538–542 131. Papinska AM, Mordwinkin NM, Meeks CJ, Jadhav SS, Rodgers
118. Munoz MC, Giani JF, Dominici FP (2010) Angiotensin-(1–7) KE (2015) Angiotensin-(1–7) administration benefits cardiac,
stimulates the phosphorylation of Akt in rat extracardiac tissues renal and progenitor cell function in db/db mice. Br J Pharmacol
in vivo via receptor Mas. Regul Pept 161:1–7. https://​doi.​org/​10.​ 172:4443–4453. https://​doi.​org/​10.​1111/​bph.​13225
1016/j.​regpep.​2010.​02.​001 132. Papinska AM, Soto M, Meeks CJ, Rodgers KE (2016) Long-term
119. Murase T, Hattori T, Ohtake M, Abe M, Amakusa Y, Takatsu administration of angiotensin (1–7) prevents heart and lung dys-
M, Murohara T, Nagata K (2012) Cardiac remodeling and dias- function in a mouse model of type 2 diabetes (db/db) by reduc-
tolic dysfunction in DahlS.Z-Lepr(fa)/Lepr(fa) rats: a new ani- ing oxidative stress, inflammation and pathological remodeling.
mal model of metabolic syndrome. Hypertens Res 35:186–193. Pharmacol Res 107:372–380. https://​doi.​org/​10.​1016/j.​phrs.​
https://​doi.​org/​10.​1038/​hr.​2011.​157 2016.​02.​026
120. Naito T, Ma LJ, Yang HC, Zuo YQ, Tang YW, Han JY, Kon V, 133. Parving HH, Brenner BM, McMurray JJ, de Zeeuw D, Haffner
Fogo AB (2010) Angiotensin type 2 receptor actions contribute SM, Solomon SD, Chaturvedi N, Persson F, Desai AS, Nico-
to angiotensin type 1 receptor blocker effects on kidney fibrosis. laides M, Richard A, Xiang Z, Brunel P, Pfeffer MA (2012)
Am J Physiol-Renal Physiol 298:F683–F691. https://​doi.​org/​10.​ Cardiorenal end points in a trial of aliskiren for type 2 diabetes.
1152/​ajpre​nal.​00503.​2009 N Engl J Med 367:2204–2213. https://​doi.​org/​10.​1056/​NEJMo​
121. Nagatomo Y, Meguro T, Ito H, Koide K, Anzai T, Fukuda K, a1208​799
Ogawa S, Yoshikawa T (2014) Significance of AT1 receptor 134. Patel VB, Bodiga S, Fan D, Das SK, Wang ZC, Wang W, Basu R,
independent activation of mineralocorticoid receptor in murine Zhong JC, Kassiri Z, Oudit GY (2012) Cardioprotective effects
diabetic cardiomyopathy. PLoS One 9:e93145. https://​doi.​org/​ mediated by angiotensin II type 1 receptor blockade and enhanc-
10.​1371/​journ​al.​pone.​00931​45 ing angiotensin 1–7 in experimental heart failure in angiotensin-
122. Nakai A, Yamaguchi O, Takeda T, Higuchi Y, Hikoso S, Taniike converting enzyme 2-null mice. Hypertension 59:1195-U1303.
M, Omiya S, Mizote I, Matsumura Y, Asahi M, Nishida K, Hori https://​doi.​org/​10.​1161/​Hyper​tensi​onaha.​112.​191650
M, Mizushima N, Otsu K (2007) The role of autophagy in car- 135. Patel VB, Mori J, McLean BA, Basu R, Das SK, Ramprasath
diomyocytes in the basal state and in response to hemodynamic T, Parajuli N, Penninger JM, Grant MB, Lopaschuk GD, Oudit
stress. Nat Med 13:619–624. https://​doi.​org/​10.​1038/​nm1574 GY (2016) ACE2 deficiency worsens epicardial adipose tissue
123. Nenna A, Nappi F, Avtaar Singh SS, Sutherland FW, Di inflammation and cardiac dysfunction in response to diet-induced
Domenico F, Chello M, Spadaccio C (2015) Pharmacologic obesity. Diabetes 65:85–95. https://​doi.​org/​10.​2337/​db15-​0399
approaches against advanced glycation end products (AGEs) in (db15-0399[pii])

13
Pflügers Archiv - European Journal of Physiology

136. Pereira RI, Snell-Bergeon JK, Erickson C, Schauer IE, Bergman 149. Russo C, Jin ZZ, Homma S, Rundek T, Elkind MSV, Sacco RL,
BC, Rewers M, Maahs DM (2012) Adiponectin dysregulation Di Tullio MR (2010) Effect of diabetes and hypertension on left
and insulin resistance in type 1 diabetes. J Clin Endocrinol Metab ventricular diastolic function in a high-risk population without
97:E642–E647. https://​doi.​org/​10.​1210/​jc.​2011-​2542 evidence of heart disease. Eur J Heart Fail 12:454–461. https://​
137. Petersen KF, Dufour S, Befroy D, Garcia R, Shulman GI (2004) doi.​org/​10.​1093/​eurjhf/​hfq022
Impaired mitochondrial activity in the insulin-resistant offspring 150. Saito T, Asai K, Sato S, Hayashi M, Adachi A, Sasaki Y, Takano
of patients with type 2 diabetes. N Engl J Med 350:664–671. H, Mizuno K, Shimizu W (2016) Autophagic vacuoles in car-
https://​doi.​org/​10.​1056/​NEJMo​a0313​14.​350/7/​664[pii] diomyocytes of dilated cardiomyopathy with initially decom-
138. Pitocco D, Tesauro M, Alessandro R, Ghirlanda G, Cardillo C pensated heart failure predict improved prognosis. Autophagy
(2013) Oxidative stress in diabetes: implications for vascular and 12:579–587. https://​doi.​org/​10.​1080/​15548​627.​2016.​11453​26
other complications. Int J Mol Sci 14:21525–21550. https://​doi.​ 151. Sampaio WO, de Castro CH, Santos RAS, Schiffrin EL, Touyz
org/​10.​3390/​ijms1​41121​525 RM (2007) Angiotensin-(1–7) counterregulates angiotensin II
139. Pittaluga M, Sgadari A, Dimauro I, Tavazzi B, Parisi P, Caporossi signaling in human endothelial cells. Hypertension 50:1093–
D (2015) Physical exercise and redox balance in type 2 diabetics: 1098. https://​doi.​org/​10.​1161/​Hyper​tensi​onaha.​106.​084848
effects of moderate training on biomarkers of oxidative stress 152. Sampaio WO, dos Santos RAS, Faria-Silva R, Machado LTD,
and DNA damage evaluated through comet assay. Oxidative Schiffrin EL, Touyz RM (2007) Angiotensin-(1–7) through
Medicine and Cellular Longevity. Artn 981242. https://​doi.​org/​ receptor Mas mediates endothelial nitric oxide synthase activa-
10.​1155/​2015/​981242 tion via Akt-dependent pathways. Hypertension 49:185–192.
140. Porrello ER, D’Amore A, Curl CL, Allen AM, Harrap SB, https://​doi.​org/​10.​1161/​01.​Hyp.​00002​51865.​35728.​2f
Thomas WG, Delbridge LM (2009) Angiotensin II type 2 recep- 153. Santos RA (2014) Angiotensin-(1–7). Hypertension 63:1138–
tor antagonizes angiotensin II type 1 receptor-mediated cardio- 1147. https://d​ oi.o​ rg/1​ 0.1​ 161/H
​ YPERT
​ ENSIO​ NAHA.1​ 13.0​ 1274
myocyte autophagy. Hypertension 53:1032–1040. https://d​ oi.o​ rg/​ 154. Santos RAS, Ferreira AJ, Verano-Braga T, Bader M (2013) Angi-
10.1​ 161/H
​ YPERT ​ ENSIO ​ NAHA.1​ 08.1​ 28488 (HYPERTENSIO otensin-converting enzyme 2, angiotensin-(1–7) and Mas: new
NAHA.108.128488[pii]) players of the renin-angiotensin system. J Endocrinol 216:R1–
141. Porrello ER, Delbridge LM (2009) Cardiomyocyte autophagy R17. https://​doi.​org/​10.​1530/​Joe-​12-​0341
is regulated by angiotensin II type 1 and type 2 receptors. 155. Satoh M, Fujimoto S, Arakawa S, Yada T, Namikoshi T, Haruna
Autophagy 5:1215–1216. https://​d oi.​o rg/​1 0.​4 161/​a uto.5.​8 .​ Y, Horike H, Sasaki T, Kashihara N (2008) Angiotensin II type
10153 1 receptor blocker ameliorates uncoupled endothelial nitric oxide
142. Privratsky JR, Wold LE, Sowers JR, Quinn MT, Ren J (2003) synthase in rats with experimental diabetic nephropathy. Neph-
AT1 blockade prevents glucose-induced cardiac dysfunction in rol Dial Transplant 23:3806–3813. https://​doi.​org/​10.​1093/​ndt/​
ventricular myocytes: role of the AT1 receptor and NADPH oxi- gfn357
dase. Hypertension 42:206–212. https://d​ oi.o​ rg/1​ 0.1​ 161/0​ 1.H​ YP.​ 156. Savini I, Catani MV, Evangelista D, Gasperi V, Avigliano L
00000​82814.​62655.​85 (01.HYP.0000082814.62655.85[pii]) (2013) Obesity-associated oxidative stress: strategies finalized
143. Putnam K, Shoemaker R, Yiannikouris F, Cassis LA (2012) to improve redox state. Int J Mol Sci 14:10497–10538. https://​
The renin-angiotensin system: a target of and contributor to doi.​org/​10.​3390/​ijms1​40510​497
dyslipidemias, altered glucose homeostasis, and hypertension 157. Schannwell CM, Schneppenheim M, Perings S, Plehn G, Strauer
of the metabolic syndrome. Am J Physiol-Heart Circ Physiol BE (2002) Left ventricular diastolic dysfunction as an early
302:H1219–H1230. https://d​ oi.o​ rg/1​ 0.1​ 152/a​ jphea​ rt.0​ 0796.2​ 011 manifestation of diabetic cardiomyopathy. Cardiology 98:33–39
144. Qi Y, Xu Z, Zhu Q, Thomas C, Kumar R, Feng H, Dostal DE, (64682[pii]64682)
White MF, Baker KM, Guo S (2013) Myocardial loss of IRS1 158. Sciarretta S, Maejima Y, Zablocki D, Sadoshima J (2018) The
and IRS2 causes heart failure and is controlled by p38alpha role of autophagy in the heart. Annu Rev Physiol 80:1–26.
MAPK during insulin resistance. Diabetes 62:3887–3900. https://​doi.​org/​10.​1146/​annur​ev-​physi​ol-​021317-​121427
https://​doi.​org/​10.​2337/​db13-​0095 159. Shah GN, Morofuji Y, Banks WA, Price TO (2013) High glucose-
145. Rajesh M, Mukhopadhyay P, Batkai S, Patel V, Saito K, Matsu- induced mitochondrial respiration and reactive oxygen species in
moto S, Kashiwaya Y, Horvath B, Mukhopadhyay B, Becker L, mouse cerebral pericytes is reversed by pharmacological inhibition
Hasko G, Liaudet L, Wink DA, Veves A, Mechoulam R, Pacher of mitochondrial carbonic anhydrases: Implications for cerebral
P (2010) Cannabidiol attenuates cardiac dysfunction, oxidative microvascular disease in diabetes. Biochem Biophys Res Commun
stress, fibrosis, and inflammatory and cell death signaling path- 440:354–358. https://​doi.​org/​10.​1016/j.​bbrc.​2013.​09.​086
ways in diabetic cardiomyopathy. J Am Coll Cardiol 56:2115– 160. Shahveisi K, Mousavi SH, Hosseini M, Rad AK, Jalali SA,
2125. https://​doi.​org/​10.​1016/j.​jacc.​2010.​07.​033 Rajaei Z, Sadeghnia HR, Hadjzadeh MAR (2014) The role of
146. Rees K, Hartley L, Day C, Flowers N, Clarke A, Stranges S local renin-angiotensin system on high glucose-induced cell tox-
(2013) Selenium supplementation for the primary prevention of icity, apoptosis and reactive oxygen species production in PC12
cardiovascular disease (Review). Cochrane Database of System- cells. Iran J Basic Med Sci 17:613–621
atic Reviews. Artn Cd009671. https://d​ oi.o​ rg/1​ 0.1​ 002/1​ 46518​ 58.​ 161. Shirakabe A, Zhai PY, Ikeda Y, Saito T, Maejima Y, Hsu CP,
Cd009​671.​Pub2 Nomura M, Egashira K, Levine B, Sadoshima J (2016) Drp1-
147. Riehle C, Wende AR, Sena S, Pires KM, Pereira RO, Zhu Y, dependent mitochondrial autophagy plays a protective role
Bugger H, Frank D, Bevins J, Chen D, Perry CN, Dong XC, Val- against pressure overload-induced mitochondrial dysfunction
dez S, Rech M, Sheng X, Weimer BC, Gottlieb RA, White MF, and heart failure. Circulation 133:1249–1263. https://​doi.​org/​
Abel ED (2013) Insulin receptor substrate signaling suppresses 10.​1161/​Circu​latio​naha.​115.​020502
neonatal autophagy in the heart. J Clin Invest 123:5319–5333. 162. Singh K, Singh T, Sharma PL (2011) Beneficial effects of angi-
https://​doi.​org/​10.​1172/​JCI71​171 (71171[pii]) otensin (1–7) in diabetic rats with cardiomyopathy. Ther Adv
148. Ristow M, Zarse K, Oberbach A, Kloting N, Birringer M, Kiehn- Cardiovasc Dis 5:159–167. https://​doi.​org/​10.​1177/​17539​44711​
topf M, Stumvoll M, Kahn CR, Bluher M (2009) Antioxidants 409281
prevent health-promoting effects of physical exercise in humans. 163. Singh VP, Bali A, Singh N, Jaggi AS (2014) Advanced glyca-
Proc Natl Acad Sci USA 106:8665–8670. https://​doi.​org/​10.​ tion end products and diabetic complications. Korean J Physiol
1073/​pnas.​09034​85106 Pharmacol 18:1–14. https://​doi.​org/​10.​4196/​kjpp.​2014.​18.1.1

13
Pflügers Archiv - European Journal of Physiology

164. Singh VP, Le B, Khode R, Baker KM, Kumar R (2008) Intracel- 175. Sun Y, Weber KT (1996) Angiotensin converting enzyme and
lular angiotensin II production in diabetic rats is correlated with myofibroblasts during tissue repair in the rat heart. J Mol Cell
cardiomyocyte apoptosis, oxidative stress, and cardiac fibrosis. Cardiol 28:851–858. https://​doi.​org/​10.​1006/​jmcc.​1996.​0080
Diabetes 57:3297–3306. https://​doi.​org/​10.​2337/​db08-​0805 176. Symeonides P, Koulouris S, Vratsista E, Triantafyllou K, Ioan-
165. Soetikno V, Sari FR, Sukumaran V, Lakshmanan AP, Harima M, nidis G, Thalassinos N, Katritsis D (2007) Both ramipril and
Suzuki K, Kawachi H, Watanabe K (2013) Curcumin decreases telmisartan reverse indices of early diabetic cardiomyopathy: a
renal triglyceride accumulation through AMPK-SREBP signal- comparative study. Eur J Echocardiogr 8:480–486. https://​doi.​
ing pathway in streptozotocin-induced type 1 diabetic rats. J Nutr org/​10.​1016/j.​euje.​2006.​09.​005
Biochem 24:796–802. https://​doi.​org/​10.​1016/j.​jnutb​io.​2012.​04.​ 177. Takahashi H, Yoshika M, Komiyama Y, Nishimura M (2011)
013 The central mechanism underlying hypertension: a review of
166. Soetikno V, Sari FR, Sukumaran V, Lakshmanan AP, Mito S, the roles of sodium ions, epithelial sodium channels, the renin-
Harima M, Thandavarayan RA, Suzuki K, Nagata M, Takagi R, angiotensin-aldosterone system, oxidative stress and endogenous
Watanabe K (2012) Curcumin prevents diabetic cardiomyopathy digitalis in the brain. Hypertens Res 34:1147–1160. https://​doi.​
in streptozotocin-induced diabetic rats: possible involvement of org/​10.​1038/​hr.​2011.​105
PKC-MAPK signaling pathway. Eur J Pharm Sci 47:604–614. 178. Takeda M, Yamamoto K, Takemura Y, Takeshita H, Hongyo K,
https://​doi.​org/​10.​1016/j.​ejps.​2012.​04.​018 Kawai T, Hanasaki-Yamamoto H, Oguro R, Takami Y, Tatara Y,
167. Stas S, Whaley-Connell A, Habibi J, Appesh L, Hayden MR, Takeya Y, Sugimoto K, Kamide K, Ohishi M, Rakugi H (2013)
Karuparthi PR, Qazi M, Morris EM, Cooper SA, Link CD, Loss of ACE2 exaggerates high-calorie diet induced insulin
Stump C, Hay M, Ferrario C, Sowers JR (2007) Mineralocorti- resistance by reduction of GLUT4 in mice. Diabetes 62:223–233.
coid receptor blockade attenuates chronic overexpression of the https://​doi.​org/​10.​2337/​db12-​0177
renin-angiotensin-aldosterone system stimulation of reduced 179. Tassone EJ, Sciacqua A, Andreozzi F, Presta I, Perticone M, Car-
nicotinamide adenine dinucleotide phosphate oxidase and cardiac nevale D, Casaburo M, Hribal ML, Sesti G, Perticone F (2013)
remodeling. Endocrinology 148:3773–3780. https://​doi.o​ rg/​10.​ Angiotensin (1–7) counteracts the negative effect of angiotensin
1210/​en.​2006-​1691 II on insulin signalling in HUVECs. Cardiovasc Res 99:129–136.
168. Stiefel P, Vallejo-Vaz AJ, Garcia Morillo S, Villar J (2011) Role https://​doi.​org/​10.​1093/​cvr/​cvt065 (cvt065[pii])
of the renin-angiotensin system and aldosterone on cardiometa- 180. Teshima Y, Takahashi N, Nishio S, Saito S, Kondo H, Fukui A,
bolic syndrome. Int J Hypertens 2011:685238. https://d​ oi.o​ rg/1​ 0.​ Aoki K, Yufu K, Nakagawa M, Saikawa T (2014) Production of
4061/​2011/​685238 reactive oxygen species in the diabetic heart - roles of mitochon-
169. Straznicky NE, Lambert EA, Nestel PJ, McGrane MT, Dawood dria and NADPH oxidase -. Circ J 78:300–306. https://​doi.​org/​
T, Schlaich MP, Masuo K, Eikelis N, de Courten B, Mariani JA, 10.​1253/​circj.​CJ-​13-​1187
Esler MD, Socratous F, Chopra R, Sari CI, Paul E, Lambert GW 181. Tetzner A, Gebolys K, Meinert C, Klein S, Uhlich A, Trebicka
(2010) Sympathetic neural adaptation to hypocaloric diet with or J, Villacanas O, Walther T (2016) G-protein-coupled receptor
without exercise training in obese metabolic syndrome subjects. MrgD is a receptor for angiotensin-(1–7) involving adenylyl
Diabetes 59:71–79. https://​doi.​org/​10.​2337/​db09-​0934 cyclase, cAMP, and phosphokinase A. Hypertension 68:185–
170. Sukumaran V, Veeraveedu PT, Gurusamy N, Yamaguchi K, Lak- 194. https://​doi.​org/​10.​1161/​Hyper​tensi​onaha.​116.​07572
shmanan AP, Ma M, Suzuki K, Kodama M, Watanabe K (2011) 182. Than A, Leow MKS, Chen P (2013) Control of adipogenesis by
Cardioprotective effects of telmisartan against heart failure in the autocrine interplays between angiotensin 1–7/Mas receptor
rats induced by experimental autoimmune myocarditis through and angiotensin II/AT(1) receptor signaling pathways. J Biol
the modulation of angiotensin-converting enzyme-2/angiotensin Chem 288:15520–15531. https://​doi.​org/​10.​1074/​jbc.​M113.​
1–7/mas receptor axis. Int J Biol Sci 7:1077–1092. https://​doi.​ 459792
org/​10.​7150/​ijbs.7.​1077 183. Thomas CM, Yong QC, Seqqat R, Chandel N, Feldman DL,
171. Sukumaran V, Veeraveedu PT, Gurusamy N, Lakshmanan AP, Baker KM, Kumar R (2013) Direct renin inhibition prevents car-
Yamaguchi K, Ma M, Suzuki K, Nagata M, Takagi R, Kodama diac dysfunction in a diabetic mouse model: comparison with
M, Watanabe K (2012) Olmesartan attenuates the development an angiotensin receptor antagonist and angiotensin-converting
of heart failure after experimental autoimmune myocarditis in enzyme inhibitor. Clin Sci (Lond) 124:529–541. https://​doi.​org/​
rats through the modulation of ANG 1–7 mas receptor. Mol Cell 10.​1042/​CS201​20448
Endocrinol 351:208–219. https://d​ oi.o​ rg/1​ 0.1​ 016/j.m
​ ce.2​ 011.1​ 2.​ 184. Tsutsui H, Kinugawa S, Matsushima S (2008) Oxidative stress
010 and mitochondrial DNA damage in heart failure. Circ J 72 Suppl
172. Sukumaran V, Watanabe K, Veeraveedu PT, Gurusamy N, Ma A:A31–37. JST.JSTAGE/circj/CJ-08–0014 [pii]
M, Thandavarayan RA, Lakshmanan AP, Yamaguchi K, Suzuki 185. Tsutsui H, Kinugawa S, Matsushima S, Yokota T (2011) Oxida-
K, Kodama M (2011) Olmesartan, an AT1 antagonist, attenu- tive stress in cardiac and skeletal muscle dysfunction associated
ates oxidative stress, endoplasmic reticulum stress and cardiac with diabetes mellitus. J Clin Biochem Nutr 48:68–71. https://​
inflammatory mediators in rats with heart failure induced by doi.​org/​10.​3164/​jcbn.​11-​012FR
experimental autoimmune myocarditis. Int J Biol Sci 7:154–167 186. Tziakas DN, Chalikias GK, Kaski JC (2005) Epidemiology
173. Sukumaran V, Watanabe K, Veeraveedu PT, Ma ML, Gurusamy of the diabetic heart. Coron Artery Dis 16(Suppl 1):S3–S10
N, Rajavel V, Suzuki K, Yamaguchi K, Kodama M, Aizawa Y (00019501-200511001-00002[pii])
(2011) Telmisartan ameliorates experimental autoimmune myo- 187. Van Gaal LF, Maggioni AP (2014) Overweight, obesity, and
carditis associated with inhibition of inflammation and oxidative outcomes: fat mass and beyond. Lancet 383:935–936. https://​
stress. Eur J Pharmacol 652:126–135. https://​doi.​org/​10.​1016/j.​ doi.​org/​10.​1016/​S0140-​6736(13)​62076-0
ejphar.​2010.​10.​081 188. van Thiel BS, van der Pluijm I, Riet LT, Essers J, Danser AHJ
174. Sun M, Dawood F, Wen WH, Chen M, Dixon I, Kirshenbaum (2015) The renin-angiotensin system and its involvement in
LA, Liu PP (2004) Excessive tumor necrosis factor activation vascular disease. Eur J Pharmacol 763:3–14. https://​doi.​org/​10.​
after infarction contributes to susceptibility of myocardial rup- 1016/j.​ejphar.​2015.​03.​090
ture and left ventricular dysfunction. Circulation 110:3221–3228. 189. Varga ZV, Giricz Z, Liaudet L, Hasko G, Ferdinandy P, Pacher P
https://​doi.​org/​10.​1161/​01.​CIR.​00001​47233.​10318.​23 (01.CIR. (2015) Interplay of oxidative, nitrosative/nitrative stress, inflam-
0000147233.10318.23[pii]) mation, cell death and autophagy in diabetic cardiomyopathy.

13
Pflügers Archiv - European Journal of Physiology

Biochim Biophys Acta 1852:232–242. https://​doi.​org/​10.​1016/j.​ 202. Yancy CW, Jessup M, Bozkurt B, Butler J, Casey DE, Drazner
bbadis.​2014.​06.​030 (S0925-4439(14)00207-5[pii]) MH, Fonarow GC, Geraci SA, Horwich T, Januzzi JL, Johnson
190. Villela D, Leonhardt J, Patel N, Joseph J, Kirsch S, Hallberg MR, Kasper EK, Levy WC, Masoudi FA, McBride PE, McMur-
A, Unger T, Bader M, Santos RA, Sumners C, Steckelings UM ray JJV, Mitchell JE, Peterson PN, Riegel B, Sam F, Stevenson
(2015) Angiotensin type 2 receptor (AT2R) and receptor Mas: a LW, Tang WHW, Tsai EJ, Wilkoff BL, Anderson JL, Jacobs AK,
complex liaison. Clin Sci (Lond) 128:227–234. https://​doi.​org/​ Halperin JL, Albert NM, Bozkurt B, Brindis RG, Creager MA,
10.​1042/​CS201​30515 (CS20130515[pii]) Curtis LH, DeMets D, Guyton RA, Hochman JS, Kovacs RJ,
191. Waddingham MT, Edgley AJ, Tsuchimochi H, Kelly DJ, Shirai Kushner FG, Ohman EM, Pressler SJ, Sellke FW, Shen WK,
M, Pearson JT (2015) Contractile apparatus dysfunction early in Stevenson WG, Commi W, Force AAT (2013) 2013 ACCF/AHA
the pathophysiology of diabetic cardiomyopathy. World J Dia- Guideline for the Management of Heart Failure A Report of the
betes 6:943–960. https://​doi.​org/​10.​4239/​wjd.​v6.​i7.​943 American College of Cardiology Foundation/American Heart
192. Wang L, Lu K, Hao H, Li X, Wang J, Wang K, Yan Z, Zhang S, Association Task Force on Practice Guidelines. J Am Coll Car-
Du Y, Liu H (2013) Decreased autophagy in rat heart induced diol 62:E147–E239. https://​doi.​org/​10.​1016/j.​jacc.​2013.​05.​019
by anti-beta1-adrenergic receptor autoantibodies contributes 203. Yaras N, Bilginoglu A, Vassort G, Turan B (2007) Restoration of
to the decline in mitochondrial membrane potential. PLoS diabetes-induced abnormal local Ca2+ release in cardiomyocytes
ONE 8:e81296. https://​doi.​org/​10.​1371/​journ​al.​pone.​00812​96 by angiotensin II receptor blockade. Am J Physiol Heart Circ
(PONE-D-12-29873[pii]) Physiol 292:H912-920. https://​doi.​org/​10.​1152/​ajphe​art.​00824.​
193. Watanabe R, Suzuki J, Wakayama K, Kumagai H, Ikeda Y, 2006
Akazawa H, Komuro I, Isobe M (2016) Angiotensin II recep- 204. Yayama K, Okamoto H (2008) Angiotensin II-induced vasodila-
tor blocker irbesartan attenuates cardiac dysfunction induced by tion via type 2 receptor: role of bradykinin and nitric oxide. Int
myocardial infarction in the presence of renal failure. Hypertens Immunopharmacol 8:312–318. https://​doi.​org/​10.​1016/j.​intimp.​
Res 39:237–244. https://​doi.​org/​10.​1038/​hr.​2015.​141 2007.​06.​012
194. Watanabe Y, Nagai Y, Takatsu K (2013) Activation and regu- 205. Ye YZ, Li J, Yuan ZX (2013) Effect of antioxidant vitamin sup-
lation of the pattern recognition receptors in obesity-induced plementation on cardiovascular outcomes: a meta-analysis of ran-
adipose tissue inflammation and insulin resistance. Nutrients domized controlled trials. Plos One 8. ARTN e56803. https://d​ oi.​
5:3757–3778. https://​doi.​org/​10.​3390/​nu509​3757 org/​10.​1371/​journ​al.​pone.​00568​03
195. Wei Y, Whaley-Connell AT, Chen K, Habibi J, Uptergrove GM, 206. Yim HE, Yoo KH (2008) Renin-angiotensin system - consid-
Clark SE, Stump CS, Ferrario CM, Sowers JR (2007) NADPH erations for hypertension and kidney. Electrolyte Blood Press
oxidase contributes to vascular inflammation, insulin resistance, 6:42–50. https://​doi.​org/​10.​5049/​EBP.​2008.6.​1.​42
and remodeling in the transgenic (mRen2) rat. Hypertension 207. Yokoyama T, Nakano M, Bednarczyk JL, McIntyre BW, Entman
50:384–391. https://​doi.​org/​10.​1161/​HYPER​TENSI​ONAHA.​ M, Mann DL (1997) Tumor necrosis factor-alpha provokes a
107.​089284 hypertrophic growth response in adult cardiac myocytes. Circula-
196. Westermann D, Rutschow S, Jager S, Linderer A, Anker S, tion 95:1247–1252
Riad A, Unger T, Schultheiss HP, Pauschinger M, Tschope 208. Yong QC, Thomas CM, Seqqat R, Chandel N, Baker KM, Kumar
C (2007) Contributions of inflammation and cardiac matrix R (2013) Angiotensin type 1a receptor-deficient mice develop
metalloproteinase activity to cardiac failure in diabetic car- diabetes-induced cardiac dysfunction, which is prevented by
diomyopathy: the role of angiotensin type 1 receptor antago- renin-angiotensin system inhibitors. Cardiovasc Diabetol 12:169.
nism. Diabetes 56:641–646. https://​doi.​org/​10.​2337/​db06-​1163 https://​doi.​org/​10.​1186/​1475-​2840-​12-​169
(56/3/641[pii]) 209. Younce CW, Kolattukudy PE (2010) MCP-1 causes cardiomyo-
197. Westermann D, Rutschow S, Van Linthout S, Linderer A, Bucker- blast death via autophagy resulting from ER stress caused by
Gartner C, Sobirey M, Riad A, Pauschinger M, Schultheiss HP, oxidative stress generated by inducing a novel zinc-finger protein,
Tschope C (2006) Inhibition of p38 mitogen-activated protein MCPIP. Biochem J 426:43–53. https://​doi.​org/​10.​1042/​Bj200​
kinase attenuates left ventricular dysfunction by mediating pro- 90976
inflammatory cardiac cytokine levels in a mouse model of dia- 210. Yu XY, Chen HM, Liang JL, Lin QX, Tan HH, Fu YH, Liu XY,
betes mellitus. Diabetologia 49:2507–2513. https://​doi.​org/​10.​ Shan ZX, Li XH, Yang HZ, Yang M, Li Y, Lin SG (2011) Hyper-
1007/​s00125-​006-​0385-2 glycemic myocardial damage is mediated by proinflammatory
198. Willemsen JM, Westerink JW, Dallinga-Thie GM, van Zonneveld cytokine: macrophage migration inhibitory factor. PLoS ONE
AJ, Gaillard CA, Rabelink TJ, de Koning EJP (2007) Angioten- 6:e16239. https://​doi.​org/​10.​1371/​journ​al.​pone.​00162​39
sin II type 1 receptor blockade improves hyperglycemia-induced 211. Yusuf S, Sleight P, Pogue J, Bosch J, Davies R, Dagenais G
endothelial dysfunction and reduces proinflammatory cytokine (2000) Effects of an angiotensin-converting-enzyme inhibitor,
release from leukocytes. J Cardiovasc Pharmacol 49:6–12. ramipril, on cardiovascular events in high-risk patients. N Engl
https://​doi.​org/​10.​1097/​Fjc.​0b013​e3180​2b31a7 J Med 342:145–153. https://d​ oi.o​ rg/1​ 0.1​ 056/N
​ EJM20​ 00012​ 0342​
199. Xie ZL, He CY, Zou MH (2011) AMP-activated protein kinase 0301
modulates cardiac autophagy in diabetic cardiomyopathy. 212. Zhang D, Shadrin IY, Lam J, Xian HQ, Snodgrass HR, Bursac N
Autophagy 7:1254–1255. https://​doi.​org/​10.​4161/​auto.7.​10.​ (2013) Tissue-engineered cardiac patch for advanced functional
16740 maturation of human ESC-derived cardiomyocytes. Biomaterials
200. Xu JJ, Lei SQ, Liu YN, Gao X, Irwin MG, Xia ZY, Hei ZQ, 34:5813–5820. https://​doi.​org/​10.​1016/j.​bioma​teria​ls.​2013.​04.​
Gan XL, Wang TT, Xia ZY (2013) Antioxidant N-acetylcysteine 026
attenuates the reduction of Brg1 protein expression in the myo- 213. Zhang F, Hu YH, Xu QB, Ye S (2010) Different effects of angio-
cardium of type 1 diabetic rats. J Diabetes Res. Artn 716219. tensin ii and angiotensin-(1-7) on vascular smooth muscle cell
https://​doi.​org/​10.​1155/​2013/​716219 proliferation and migration. Plos One 5. ARTN e12323. https://​
201. Xu XM, Kobayashi S, Chen K, Timm D, Volden P, Huang Y, doi.​org/​10.​1371/​journ​al.​pone.​00123​23
Gulick J, Yue ZY, Robbins J, Epstein PN, Liang QR (2013) 214. Zhao W, Ahokas RA, Weber KT, Sun Y (2006) ANG II-induced
Diminished autophagy limits cardiac injury in mouse models of cardiac molecular and cellular events: role of aldosterone. Am
type 1 diabetes. J Biol Chem 288:18077–18092. https://​doi.​org/​ J Physiol Heart Circ Physiol 291:H336-343. https://​doi.​org/​10.​
10.​1074/​jbc.​M113.​474650 1152/​ajphe​art.​01307.​2005

13
Pflügers Archiv - European Journal of Physiology

215. Zhao W, Li YL, Jia LX, Pan LL, Li HH, Du J (2014) Atg5 defi- 218. Zuanetti G, Latini R, Maggioni AP, Franzosi M, Santoro L, Tog-
ciency-mediated mitophagy aggravates cardiac inflammation noni G (1997) Effect of the ACE inhibitor lisinopril on mortality
and injury in response to angiotensin II. Free Radical Biol Med in diabetic patients with acute myocardial infarction: data from
69:108–115. https://d​ oi.o​ rg/1​ 0.1​ 016/j.f​ reera​ dbiom
​ ed.2​ 014.0​ 1.0​ 02 the GISSI-3 study. Circulation 96:4239–4245. https://d​ oi.o​ rg/1​ 0.​
216. Zhou G, Li X, Hein DW, Xiang X, Marshall JP, Prabhu SD, 1161/​01.​cir.​96.​12.​4239
Cai L (2008) Metallothionein suppresses angiotensin II-induced
nicotinamide adenine dinucleotide phosphate oxidase activation, Publisher's Note Springer Nature remains neutral with regard to
nitrosative stress, apoptosis, and pathological remodeling in the jurisdictional claims in published maps and institutional affiliations.
diabetic heart. J Am Coll Cardiol 52:655–666. https://d​ oi.o​ rg/1​ 0.​
1016/j.​jacc.​2008.​05.​019
217. Zhou MS, Schulman IH, Zeng Q (2012) Link between the renin-
angiotensin system and insulin resistance: Implications for car-
diovascular disease. Vasc Med 17:330–341. https://​doi.​org/​10.​
1177/​13588​63X12​450094

13

You might also like