Sei sulla pagina 1di 13

Nutrition, Metabolism & Cardiovascular Diseases (2017) 27, 657e669

Available online at www.sciencedirect.com

Nutrition, Metabolism & Cardiovascular Diseases


journal homepage: www.elsevier.com/locate/nmcd

REVIEW

Metformin effects on the heart and the cardiovascular system:


A review of experimental and clinical data
L. Nesti, A. Natali*
Department of Clinical and Experimental Medicine, University of Pisa, Italy

Received 9 February 2017; received in revised form 12 April 2017; accepted 21 April 2017
Available online 10 May 2017

KEYWORDS Abstract Background: Metformin, the eldest and most widely used glucose lowering drug, is
Metformin; likely to be effective also on cardiac and vascular disease prevention. Nonetheless, uncertainty
Heart; still exists with regard to its effects on the cardiovascular system as a whole and specifically
Cardiovascular on the myocardium, both at the organ and cellular levels.
Methods: We reviewed the available data on the cardiac and vascular effects of metformin, en-
compassing both in vitro, either tissue or isolated organ, and in vivo studies in experimental an-
imals and humans, as well as the evidence generated by major clinical trials.
Results: At the cellular level metformin’s produces both AMP-activated kinase (AMPK) dependent
and independent effects. At the systemic level, possibly also through other pathways, this drug
improves endothelial function, protects from oxidative stress and inflammation, and from the
negative effects of angiotensin II. On the myocardium it attenuates ischemia-reperfusion injury
and prevents adverse remodeling induced by humoral and hemodynamic factors. The effects on
myocardial cell metabolism and contractile function being not evident at rest or in more
advanced stages of cardiac dysfunction, could be relevant during transient ischemia, during an
acute increase in workload and in the early stages of diabetic/hypertensive cardiomyopathy as
confirmed by few small clinical trials and some observational studies. The overall evidence
emerging from both clinical trials and real world registry is in favor of a protective effect of met-
formin with respect to both coronary events and progression to heart failure.
Conclusions: Given this potential, its efficacy and its safety (and also its low cost) metformin re-
mains the central pillar of the therapy of diabetes.
ª 2017 The Italian Society of Diabetology, the Italian Society for the Study of Atherosclerosis, the
Italian Society of Human Nutrition, and the Department of Clinical Medicine and Surgery, Feder-
ico II University. Published by Elsevier B.V. All rights reserved.

Introduction with any other blood glucose-lowering drugs. Originally


derived from galegine (isoamylene guanidine), a guanidine
Metformin, a biguanide derivative (dimethylbiguanide), is derivative found in the French lilac Galega officinalis, more
worldwide the most used drug for the treatment of diabetes than 50 years ago, this drug has an incredible enduring re-
mellitus type 2 (DM2), being recommended by major cord. The hypoglycemic effect of metformin derives from an
guidelines both as first-line therapy and in combination insulin-sensitizing action both on the liver, resulting in a
reduced hepatic glucose release, and, to a minor extent, at
the level of peripheral tissues, where it promotes glucose
* Corresponding author. Department of Clinical and Experimental
uptake [1]. Together with intestinal disturbances, the major
Medicine, University of Pisa, Via Savi 8, 56100 Pisa, Italy.
E-mail address: andrea.natali@med.unipi.it (A. Natali).
metformin-related adverse effect is the rise in blood lactate

http://dx.doi.org/10.1016/j.numecd.2017.04.009
0939-4753/ª 2017 The Italian Society of Diabetology, the Italian Society for the Study of Atherosclerosis, the Italian Society of Human Nutrition, and the Department of Clinical
Medicine and Surgery, Federico II University. Published by Elsevier B.V. All rights reserved.
658 L. Nesti, A. Natali

levels, for which its use has been discouraged in patients involved mechanisms and the relevance. For each topic,
with chronic kidney disease, hepatic or respiratory insuffi- we will focus first on experimental and then on clinical
ciency, and heart failure (HF). More recently, however, the studies.
evidence that metformin-related lactic acidosis has a very
low incidence even in the presence of predisposing
comorbidities [2e5], has led to a revision by the FDA and Effects of metformin on glucose and lipid cell
EMEA of the drug labeling [6], allowing its use in subjects metabolism (Fig. 1)
with estimated glomerular filtration rate values above
30 ml/min/1.73 m2 and, with some cautions, in chronic HF. Despite its long-term use, the pharmacodynamics of
Besides having few adverse effects, metformin pos- metformin has been only recently fully clarified. Its effects
sesses several additional properties beyond glycemic were initially supposed to be mediated mainly by the
control. There is research and clinical interest in using this activation of AMP-activated protein kinase (AMPK), which
drug for polycystic ovary disease [7] as well as in cancer is highly expressed not only in the liver but also in other
prevention and treatment [8,9]. A potential beneficial role tissues, including the intestine, adipose, and skeletal
for metformin has been also suggested in preeclampsia muscle and in the myocardium. More recently, however,
prevention and treatment [10] and in multiple sclerosis other AMPK-independent actions have also been
[11]. Interestingly, by mimicking some of the benefits of demonstrated.
caloric restriction, both in drosophila and in mice, met-
formin has been observed to improve healthspan and AMPK-dependent mechanisms of action
lifespan [12]. Despite the evidence from the United
Kingdom Prospective Diabetes Study (UKPDS) clinical trial Both in vitro and in vivo, metformin can activate AMPK, with
that metformin therapy in overweight newly diagnosed either direct or indirect pathways [14]. Noteworthy, the
DM2 patients is associated with a relevant reduction in metabolic effect of metformin, both after acute [15] and
cardiovascular endpoints [13], information on the cardio- chronic treatment [16], occurs without an evident increase
vascular effects of metformin is controversial, sparse, and in intracellular total AMPK levels, being therefore depen-
limited. With this review, we aim at synthetizing and dent upon the increase in the phosphorylated-to-
critically evaluating the available evidence on metformin’s unphosphorylated AMPK ratio. It directly binds to AMPK
effects on the cardiovascular system, heart functions, subunits, resulting in an increased assembly of the enzy-
structure, and metabolism, and, when possible, discuss the matically active, heterotrimeric complex, which is more

Figure 1 Effects of metformin on cell metabolism. Intracellular signaling molecules activated by metformin are shown with red continuous lines;
those being inhibited are shown with blue dotted lines. The black lines describe the consequences on other substrates or on metabolic pathways (in
italics). Blue and red boxes indicate inhibited and activated proteins, respectively. ACC1/2: Acetyl-CoA Carboxylase-1/2. AMPK: AMP-activated
protein kinase. FFA: free fatty acids. AKT: also known as protein kinase B. FOxO1: Forkhead box protein 1. GLUT4: glucose transporter 4. PCK-e:
protein kinase C-epsilon. PFKFB3: 6-phosphofrutto-2-kinase/fructose-2, 6-biphosphatase 3 kinase.
Metformin effects on the heart and the cardiovascular system 659

accessible for upstream kinases [17,18]. In addition, metfor- Moreover, insulin-sensitizing effects of metformin have
min is also capable of indirectly activating AMPK, that is by also been linked to a modification in microbiota compo-
altering AMP/ADP-to-ATP ratio in at least two ways: first, by sition [31e33]. In high-fat diet-fed and diabetic rodents, it
blocking AMP-deaminase [19], and second, by inhibiting leads to a decrease in postprandial serum gut microbiota-
mitochondrial complex 1, a transmembrane protein derived lipopolysaccharide (LPS) [34], which has been
involved in the respiratory chain [20]. The consequence is an recently recognized to have an important role in the
increase in AMP/ADP-to-ATP ratio, which is in turn development of insulin resistance and endothelial
responsible for AMPK activation. In this regard, metformin dysfunction [35]. Interestingly, metformin reduction of
mimics a condition of imbalance between energy supply and postprandial hyperglycemia has been linked to an increase
use similar to the conditions of fasting and exercise. in gut Akkermansia spp. population and attenuation in
This increase in AMPK catalytic activity affects adipose tissue inflammation by inducing Foxp3 regulatory
numerous metabolic pathways modulating some key en- T cells [36].
zymes in energy-consuming biosynthetic pathways, such
as hydroxymethylglutaryl-CoA reductase, acetyl-CoA Effects of metformin on the vascular system (Fig. 2)
carboxylase 1 and 2 (ACC1 and ACC2), and glycogen syn-
thase [21]. Interestingly, in muscle and fat tissues, AMPK Oxidative stress, inflammation and endothelial function
regulates glucose uptake through effects on the RabGAP
TBC1D1 protein, thus influencing GLUT4-mediated glucose Metformin seemingly participates in protecting against
uptake [22]. In addition to acutely regulate these enzymes, oxidative stress [37,38]. It attenuates LPS-induced [39] and
AMPK is also involved in an adaptive reprogramming of oxidized LDL-induced proinflammatory responses [40,41],
metabolism through transcriptional changes. One example suppresses macrophage proinflammatory responses
is the recently discovered inhibition of the lipogenic [39,42,43], and promotes macrophage differentiation to an
transcriptional factor Srebp1 [23] that induces ACC1 anti-inflammatory functional phenotype [44]. In a model
stimulated fatty acid synthesis. As a consequence, AMPK of fatty liver disease, metformin therapy normalizes he-
activation prompts a switch from ATP-consuming anabolic patic anaplerosis/cataplerosis and reduces the markers of
pathways to ATP-generating catabolic pathways, particu- inflammation by preventing the concomitant elevation of
larly inhibiting the synthesis of triglycerides and proteins, oxidative metabolism [45]. The decreased production of
and stimulating fatty acid oxidation, promoting glucose ROS is linked to the inhibition of PKC [46] and complex 1 of
transport, and accelerating glycolysis [24], thereby the respiratory chain [47,48], while the anti-inflammatory
reducing intracellular lipid stores. Pathological accumula- effect to the inhibition of IKKa/b [49]. Moreover, the
tion of lipids in the liver impairs insulin signaling through transcription factor SKN-1/Nrf2 is activated upon metfor-
the diacylglycerols mediated activation of protein kinase C min treatment, resulting in an increased expression of
ε (PKC-ε) [25]. The central role of intracellular lipids has antioxidant genes in cells and animal models [50]. Inter-
been proven by Fullerton et al. [26] who observed that estingly, metformin yields its anti-inflammatory effect
metformin’s hypoglycemic effect depends on its ability to irrespective of diabetic status [49].
lower cellular fatty acid levels through the AMPK- In a chronic heart transplantation model, Chin et al. [51]
dependent phosphorylation of ACC1 and ACC2. Further- demonstrated that metformin significantly reduced the
more, metformin also seems to improve glucose homeo- extent of microvascular damage, and this was associated
stasis indirectly through the so-called “gut-brain-liver axis” with improved cardiac grafts function. It has been reported
[27]. In rats, the intraduodenal metformin infusion is that in aortic endothelial cells, the activation of AMPK by
capable of reducing liver fasting glucose production by metformin limits endothelial cell damage caused by
activating duodenal enterocyte AMPK and PKA-dependent oxidative stress under hyperglycaemic conditions through
GLP-1 secretion [28]. the inhibition of the protein kinase C-NAD(P)H oxidase
pathway [52]. The resulting reduction in ROS generation
AMPK-independent mechanisms of action halts the activation of endothelial apoptosis triggered by
the loss of mitochondrial membrane potential [53]. Met-
Recently, it was demonstrated that metformin action on formin has shown a beneficial effect on nonendothelial
glucose control is maintained in AMPK/LKB1 knockout function as well, potentiating phenylephrine-induced
rodents [29], in which it inhibits hepatic gluconeogenesis AMPK phosphorylation and promoting vasorelaxation in
by decreasing the hepatocyte global energy state [30]. The endothelium-denuded rat aortic rings [54]. Furthermore,
mechanism is through the inhibition of mitochondrial both in isolated cardiomyocytes and in the beating heart, it
glycerol-3-phosphate dehydrogenase, which alters the can mitigate endothelial dysfunction by inhibiting oxida-
cytosolic redox state (as reflected by a higher lactate-to- tive stress-activated poly (ADP-ribose) polymerase 1
pyruvate ratio) and increases the mitochondrial redox (PARP1) [55]. Davis et al. [56] reported that metformin
state (as reflected by beta-hydroxybutyrrate-to- increased the phosphorylation of eNOS in an AMPK-
acetoacetate ratio) [16]. By blocking this enzyme, metfor- dependent manner, which in turn increased eNOS activ-
min reduces the NADþ available for converting lactate into ity and NO bioavailability. Interestingly, in the heart, eNOS
pyruvate, thereby reducing gluconeogenesis and sup- is expressed not only by vascular endothelial cells but also
pressing fasting hepatic glucose production. by cardiomyocytes [57]. Moreover, early treatment with
660 L. Nesti, A. Natali

Figure 2 Effects of metformin on the vasculature. Metformin can indirectly exert a protective effect on the vasculature mainly by reducing
inflammation and oxidative stress and protecting from fibrosis and remodeling. Blue and red indicate inhibition and activation, respectively. AT1R:
Angiotensin II receptor 1. (For interpretation of the references to color in this figure legend, the reader is referred to the web version of this article.)

metformin was observed to reduce pulmonary pressures positive [66] or negative [67] results. However, while the
and vascular remodeling in a rat model of pulmonary hy- former study did not control for the improvement in
pertension associated with HF with preserved ejection plasma glucose being versus placebo, the latter had an
fraction (EF), an effect that was associated with a signifi- active treatment arm.
cant activation of SIRT3 and AMPK [58]. Interestingly, in We can conclude that metformin produces a selective
rodents, metformin seems to also reverse pulmonary hy- inhibition of oxidative stress and inflammation both
pertension by inhibiting aromatase and estrogen [59], directly and indirectly. Moreover, it can reduce cellular
which in the smooth muscle cell of the pulmonary artery metabolic overload and the insulin resistance-induced
are a promitogen factor. Moreover, it can modulate pul- inflammation in several tissues and act directly on
monary vascular remodeling by reducing smooth muscle monocytes by inhibiting their differentiation to macro-
cells proliferation through the inhibition of mTOR [60]. phages. The beneficial effects of metformin on endothelial
In humans, there is consistent evidence supporting an function, evident in animals, still have to be convincingly
anti-inflammatory effect of metformin particularly in confirmed in humans. The effects observed in animal
obese women with impaired glucose tolerance (IGT) [61] models on pulmonary hypertension are of particular in-
and polycystic ovary disease [62]. In the BARI trial, the terest and deserve further research.
insulin-sensitizing strategy (mainly metformin, but also
thiazolidinediones) led to lower C-reactive protein and
fibrinogen serum concentrations at all follow-up evalua- Renin-angiotensin-aldosterone system
tions after baseline in subjects with DM2 and coronary
artery disease [63]. In a large and accurate trial versus There is some evidence suggesting that metformin might
placebo with similar glucose control [64], the positive ef- interfere with the renin-angiotensin-aldosterone system
fect of metformin extended to several plasma markers of (RAAS). The study by Lee et al. [68], performed in cultured
endothelial dysfunctions (s-ICAM, sVCAM-1, t-PA, and PAI- proximal renal tubule cells, observed metformin-induced
1) and persisted for the whole 52 weeks of the observation activation of AMPK to reduce angiotensin II (AngII) and
period. With regard to vascular endothelial function, 8 aldosterone-induced epithelialemesenchymal transition.
weeks of treatment with metformin 500 mg bid resulted In the same study, metformin reduced fibrogenesis
in an improvement in skin microcirculation, endothelial through the reduction of cytosolic reactive oxygen species
function, and decreased myocardial ischemia in nondia- (ROS) production. In addition, in fibroblasts, pretreatment
betic women with microvascular angina (positive exercise with metformin could decrease the production of extra-
test and normal coronaries) [65]. Two studies in DM2 cellular matrix and the activation of ERK signaling induced
patients focused on resistance arteries, yielding either by AngII. More interestingly, in AngII-induced hypertrophy
Metformin effects on the heart and the cardiovascular system 661

in cultured cardiomyocytes, metformin exerted anti- Effects of metformin on the heart (Fig. 3)
hypertrophic effects and prevented AngII-induced cell
death [69]. This effect was related to blunting of AngII- Several in vitro and in vivo animal studies documented
induced upregulation of AngII type 1 receptor (AT1R). some direct effects of metformin both on isolated car-
Moreover, metformin attenuated mitochondrial dysfunc- diomyocytes and on the beating heart. On the contrary,
tion and hypertrophy through the eNOS/SIRT1/p53 studies in humans are far less abundant. Compelling evi-
pathway. However, the exact mechanisms through which dence suggests that metformin plays several roles in
metformin-induced AMPK activation leads to AT1R inhi- different cardiomyocyte activities, such as electric activity,
bition in cardiomyocyte mitochondria are still to be energy metabolism, ischemia-reperfusion injury, cardiac
elucidated. It is noteworthy that metformin has been remodeling, as well as systolic and diastolic functions.
shown [70] to attenuate atherosclerosis and vascular
senescence in a high-fat diet-fed mice and to prevent the Electric potential and conduction
upregulation of AT1R by a high-fat diet in the aortas of
mice. We are not aware of data concerning the influence of Few data are available regarding metformin influence on
metformin on the RAAS in humans. cardiomyocytes electric activity. In a type 1 diabetes
In synthesis, it seems unlikely that metformin directly mouse model (diabetes induced by alloxan infusion),
modulates the RAAS. However, it might prevent the adverse chronic treatment with metformin doses effective in
effect of AngII both at the level of the myocardium (fibrosis) achieving plasma glucose control prevented the increase
and the vasculature (atherosclerosis). in QT interval and dispersion induced by the induction of
diabetes [88]. In a rat model of acute myocardial infarction
Blood pressure and sympathetic activity (MI), short-term oral administration of metformin
strongly prevented pathologic alterations in the ECG,
In mouse models, intravenous administration of metfor- indicating its protective effects on cell membrane function
min produced a dose-dependent reversible decrease in [89]. As far as humans are concerned, only one study has
mean arterial pressure, heart rate, and efferent renal been conducted, wherein DM2 patients reported a 12
sympathetic activity [71]. Other studies confirmed an msec reduction in corrected QT interval (QTc) and a
antihypertensive effect of metformin in animals. In neutral effect in QTc dispersion after 2 months of met-
particular, metformin has been proven to attenuate salt- formin treatment [90].
induced hypertension in spontaneously hypertensive rats In conclusion, there is no clear evidence of a direct,
[72], although it did not affect blood pressure in animals significant influence of metformin treatment on car-
on normal salt diet [73]. diomyocyte electric activity.
Two preliminary small studies in humans showed a
slight blood pressure lowering effect of metformin in DM2 Energy metabolism of the normal heart
patients with hypertension [74,75]. These observations
were not confirmed in more recent studies in subjects In isolated cardiomyocytes, metformin activates glucose
with DM2 [76e79] and in nondiabetic, hypertensive pa- uptake and glycolysis by both AMPK-dependent [91,92]
tients [80e83], in whom the measurement of blood pres- and AMPK-independent (p38-MAPK and PKC) [93] mech-
sure was generally more accurate. Interestingly, the effect anisms, depending on the concentration and duration of
of metformin was not additive to the effect of exercise in exposure to the drug, while the increase in fatty acid
normotensive subjects with IGT [84] and was unable to oxidation is evident only if AMPK is activated [94]. In intact
prevent the development of hypertension in IGT subjects myocardium, metformin promotes glucose transport, ac-
[61,85,86]. A recent meta-analysis, however, suggested celerates glycolysis, and stimulates fatty acid oxidation,
that metformin could effectively lower systolic blood inhibiting the synthesis of triglycerides. In particular, in
pressure in nondiabetic patients, especially in those with the normal intact heart, the effect of metformin on fatty
IGT or obesity [87], a result possibly related to the small acid utilization appears to predominate and to occur
size of the effect and of the sample of each single study. independently of AMPK activation [93]. In an in vivo DM2
With regard to autonomic function, despite one work on experimental animal model (fa/fa), however, chronic
heart rate variability suggested a positive effect for met- treatment with metformin (16 mg/k/die) was not associ-
formin in affecting cardiac autonomic tone [76], more ac- ated with a different pattern of myocardial substrates
curate studies using micro neuronography and utilization [95].
norepinephrine kinetics showed no effect of metformin on Studies in humans with positron emission tomography
the sympathetic nervous system [82]. are somewhat conflicting: while no effect on glucose up-
The overall balance of evidence indicates that metfor- take was observed by Hällsten [96], van der Meer [97]
min does not have a significant direct effect on sympa- demonstrated a small reduction in both glucose and FFA
thetic activity, while it might have some positive, though utilization, and Lyons [98] reported no direct effect on
small, effects in reducing systolic blood pressure. The ef- substrate utilization.
fect seems to be more relevant in patients with obesity Considering the data summarized above, despite the
and/or IGT, possibly secondary to the metformin-related effect observed in vitro, metformin at “normal” pharma-
body weight reduction. cologic concentrations is unlikely to produce major
662 L. Nesti, A. Natali

Figure 3 Effects of metformin on the heart. Metformin has been suggested to exert effects on electric activity, cardiomyocyte metabolism, ischemia,
remodeling, as well as on myocardial systolic and diastolic functions. For each function, we have summarized the evidence according to the type of
experiments. IGT: impaired glucose tolerance. DM: diabetes mellitus. LV: left ventricle.

changes in myocardial substrate utilization in the intact Noteworthy, the cardioprotective effect of metformin in
beating heart, under normal resting conditions. Nonethe- ischemia-reperfusion injury has been demonstrated both
less, it should be noted that in resting and fasting condi- in diabetic [105] and in nondiabetic rodent models
tions, the energy metabolism of the heart relies mainly on [100e104]. The beneficial effects of metformin on
the availability of circulating FFA, whose serum concen- ischemia-reperfusion injury have been proposed to be
tration is extremely sensitive to factors such as the dura- secondary to either a more efficient shift from fatty acid
tion of fasting, stress, fat mass, insulin, and glucagon aerobic oxidation to glucose anaerobic utilization or to an
relative serum concentrations. As such, it cannot be improvement in cardiomyocyte insulin signaling [106].
excluded that some of these confounding factors might be Finally, the cytoprotective effects of metformin were
responsible for the variability observed in the results associated with AKT phosphorylation and successive in-
in vivo. hibition of mitochondrial permeability transition pore
opening [107].
Observational studies in humans are consistent with a
Ischemia-reperfusion injury protective role for metformin in acute ischemic heart
disease, although a potential harm of other blood glucose
In a study performed in the nondiabetic isolated rat heart lowering therapy has been proposed as a confounding
model, it has been found that adding metformin to the factor. In particular, metformin treatment, when compared
perfusate resulted in the attenuation of LV postischemic to other hypoglycemic agents, was associated with
dysfunction (stunning) [99]. The beneficial effects of short- improved survival after ST-elevation MI (STEMI) in pa-
term metformin administration have been further sus- tients with DM2 [108]. More recent studies confirmed that
tained by some studies in vivo, wherein acute metformin in patients with DM2 and a history of MI, treatment with
therapy significantly reduced postischemic myocardial metformin was associated with a lower mortality rate in
damage [100]. Several studies, performed in both isolated comparison with sulfonylureas (SUDs) [13,109]. A retro-
hearts and intact animals [101e105], provided support to spective study suggested a role for metformin in reducing
the notion that metformin administration can reduce the incidence of the so-called no-reflow phenomenon in
infarct size: compared to the control group, metformin patients with DM2 after primary angioplasty for acute MI
administration before or during ischemia-reperfusion [110]. In DM2 patients with acute MI treated with met-
injury was associated with an infarct size reduction be- formin, with regard to nonmetformin-treated diabetic
tween 22% [101] and 65% [105]. The infarct size-limiting patients, lower peak values of cardiac cell death markers,
effect of metformin was demonstrated in mice after such as creatine kinase (CK), mbCK, and troponins, were
either a single intraperitoneal injection or a single intra- observed [111]. Moreover, the same study demonstrated a
ventricular administration during reperfusion. smaller infarct size in metformin-treated DM2 patients
Metformin effects on the heart and the cardiovascular system 663

than in the nondiabetic patients, not taking the drug [111]. metformin can exert cardiac protective effects through an
A similar study, however, did not confirm this finding AMPK-independent molecular pathway.
[112]. However, despite the substantial effect in diabetic Similar beneficial effects have also been proved for long-
patients, metformin treatment (500 mg bid) initiated term metformin administration in rodent models of
immediately after percutaneous transluminal coronary ischemic HF, wherein both short- (4 weeks) [122] and long-
angioplasty (PTCA) in nondiabetic patients suffering from term (12 weeks) [101] metformin treatment after MI
coronary heart disease was associated with no clear significantly attenuated cardiac remodeling and retarded
beneficial effect on NT-proBNP levels or LVEF at 4 months, the progression of ischemic cardiomyopathy and HF.
as demonstrated by the Glycometabolic Intervention as Particularly, rats treated with metformin showed reduced
Adjunct to Primary Percutaneous Coronary Intervention in MI size, improved echocardiographically assessed LV ge-
ST-Segment Elevation Myocardial Infarction (GIPS-III) trial ometry, and a smaller increase in molecular correlates of LV
[113]. No other beneficial effect has been observed in remodeling, such as ANP and collagen production. In
nondiabetic subjects suffering from coronary artery dis- addition, in models of postischemic HF after permanent left
ease [114]. Similarly, short-term metformin pretreatment coronary artery occlusion, metformin administration
was not effective in reducing peri-procedural myocardial significantly improved LV remodeling, as evidenced by in-
injury in nondiabetic patients undergoing coronary artery creases in LV systolic pressure and LVEF, as well as by de-
bypass graft surgery [115]. However, in patients with creases in LV end-diastolic diameter and LV end-systolic
metabolic syndrome, metformin treatment (250 mg, 3 diameter [102,122]. These beneficial effects of metformin
times daily) started 7 days prior to elective percutaneous were associated with increased AMPK and eNOS phos-
coronary intervention resulted in both a smaller (50%) phorylation. Of note, a genetic link between AMPK and
cardiac biomarker release after intervention and an cardiac fibrosis has been recently demonstrated in a MI
extremely favorable 1-year clinical outcome (8% vs. 29% mouse model [123]. In addition, a model of spontaneously
major clinical events) [116]. hypertensive, insulin-resistant rats [124] demonstrated
In conclusion, in nondiabetic subjects, acute adminis- that 12 months-long metformin treatment was associated
tration of the drug during or soon after an ischemic event with reduced echographically assessed LV volumes and
yields no cardioprotection. Conversely, data strongly sug- wall stress, as well as histologically proven perivascular
gest that in subjects with diabetes or with metabolic fibrosis and cardiac lipid accumulation in an AMPK-
syndrome, as well as in diabetic animal models, chronic dependent mechanism. In the same study, metformin
pretreatment with metformin exerts beneficial anti- induced a marked activation of eNOS and vascular endo-
ischemic effects, reducing ischemia-reperfusion injury, thelial growth factor, and reduced TNF-a expression and
infarct size, and cardiac cell death markers serum levels, myocyte apoptosis. Myocyte apoptosis has been also stud-
possibly justifying the improved survival after an ischemic ied after metformin administration in a swine model of
event reported in the observational studies. chronic myocardial ischemia, wherein metformin treat-
ment reduced the percentage of apoptotic cells in both
Remodeling ischemic and nonischemic myocardium [106]. This car-
dioprotective effect was due to both upregulation of pro-
Metformin reduces cardiac hypertrophy in mice subjected survival proteins, such as extracellular signal-regulated ki-
to chronic pressure overload, partially through AMPK- nases, nuclear factor kB, phosphorylated eNOS, and P38,
independent molecular pathways [117]. Metformin was and downregulation of pro-apoptotic proteins, such as
also shown to attenuate cardiac fibrosis and inhibit FOXO3 and caspase 3. In diabetic mice, cardiac autophagic
collagen synthesis in both a mouse model of pressure activities were also enhanced by metformin [125]. How-
overload-induced HF [118] and a mouse model of oxidative ever, a conclusive link between increased autophagy and
stress-induced ventricular hypertrophy, in which it can cardiac remodeling in metformin-treated diabetic hearts
significantly inhibit the overexpression of TGF-b1 and has not been demonstrated.
bFGF [119]. In a rat model, while 2 days-long isoproterenol To date, no clinical study has been conducted to
administration resulted in cardiac hypertrophy and investigate the effect of metformin on LV remodeling. The
increased edematous intramuscular space with decreased results of the ongoing MET-REMODEL trial [126], a phase
myocardial compliance, the concomitant oral administra- IV, randomized, placebo-controlled trial, aimed at inves-
tion of metformin clearly and dose-dependently prevented tigating the efficacy of metformin in regression of LV hy-
myocardial injury, edema, and massive fibrosis. Interest- pertrophy in patients with coronary artery disease and
ingly, in a canine model of tachycardia-induced HF, met- insulin resistance are eagerly awaited. The primary
formin reduced myocardial fibrosis through reduced endpoint of this trial is to investigate any change in LV
TGFb1 mRNA and could prevent the progression of HF as mass index. Secondary endpoints include changes in in-
well [120]. Nevertheless, in mice exposed to transverse sulin resistance measured using fasting insulin resistance
aortic constriction-induced hypertrophy, metformin index (FIRI), obesity, and LV size and function, thus
significantly improved LV fractional shortening, attenuated possibly clarifying metformin’s effect on remodeling in
LV dilation, and was associated with a smaller increase in humans.
serum ANP [121]. This effect was observed in both wild- In conclusion, while studies in humans are currently
type and AMPK-knockout animals, indicating that lacking, in animal models, metformin shows potent anti-
664 L. Nesti, A. Natali

remodeling properties, seemingly through reduced inter- observation found no clear effect of metformin on systolic
stitial fibrosis, reduced inflammation-related myocardial and diastolic function. In a small randomized clinical trial,
damage, and seemingly limiting myocardial hypertrophy. 24 weeks of treatment with metformin (2000 mg) had no
Interestingly, these beneficial effects are present irre- effect on diastolic function as assessed by echocardiog-
spective of either the nature of myocardial insults or the raphy [97] in patients with normal left ventricular func-
glycemic status. tion and negative dobutamine stress test. Finally, the
already cited GIPSS trial [13,114] observed no change in
Systolic and diastolic functions LVEF in diabetic patients treated with metformin. How-
ever, it should be noted that in the GIPS III study, the low
In models of postischemic HF after permanent left coro- dose of metformin used (1 g daily) and the short treat-
nary artery occlusion, metformin administration signifi- ment period of treatment (4 months) may not be enough
cantly improved LVEF [102,122] and daily survival by 47% to have an impact on the endpoint of LVEF modification.
(vs. vehicle) at 4 weeks following permanent left coronary Very recently, a clinical trial in 105 patients with DM2
artery occlusion [102]. Furthermore, 1-month-long met- without history of cardiac disease, aiming at evaluating
formin treatment was associated with preserved cardiac the effect of improving metabolic control on left ventric-
function in an in vivo rat model of posteMI cardiac ular systolic function (as measured by global longitudinal
remodeling [101]. In the setting of nonischemic HF, while strain), reported that the use of metformin was the only
in the rodent model of volume-overload HF, metformin did other independent predictor of the improvement in left
not show any effect on systolic or diastolic function [127], ventricular contractility in addition to the change in
but in streptozotocin-treated animals, long-term oral HbA1c and weight [133].
administration of metformin increased the myocardial In conclusion, only in the presence of acute stress
performance to an increase in preload [128]. The protec- (ischemia, acute preload and post preload increase) and in
tive effect of metformin in workload-dependent HF has early diabetic cardiomyopathy, metformin might positively
been shown to be dependent on the AMPK pathway [129]. affect cardiac performance.
In their impressive series of experiments in the working
rat ex vivo, Taegtmeyer and colleagues [129] showed that Metformin and major cardiac clinical outcomes
the accumulation of G6P, due to the glucose uptake/
oxidation mismatch induced by an increased workload, is Ischemic cardiovascular events prevention
responsible for downstream mTOR activation with the
consequent endoplasmatic reticulum stress and contractile Three relatively small, randomized clinical trials have
dysfunction. Metformin treatment in vivo, and added to reached similar conclusions with regard to the potential
the perfusate, by preventing the AMPK downregulation positive effects of metformin on macrovascular outcomes.
preserved the coupling between glucose uptake and In 1998, the UKPDS study [134] demonstrated that in obese
oxidation, avoided the intracellular accumulation of G6P, subjects with DM2, metformin compared to insulin or SUDs
and improved myocardial energetics at high workload. therapy, despite similar glycemic control, was associated
Similarly, as mentioned earlier, in an elegant rodent model with a reduction in 10-year overall mortality and diabetes-
of HF with preserved EF, the chronic treatment with related death by 36% and 42%, respectively, and lowered the
metformin could reduce the elevation in right ventricular risk of MI by 39%. Importantly, the beneficial effects of
pressure and the extent of pulmonary small arteries hy- metformin persisted for 10-year of post-trial follow-up [13].
pertrophy [58]. Finally, also in a canine model of DM2, A similar (40%) reduction of macrovascular events (as a
short-term metformin treatment has been shown to heterogeneous composite secondary endpoint) was
attenuate the increment of LV diastolic chamber stiffness observed over 4.3 years in the HOME trial [135], in which
and diastolic dysfunction [130]. metformin was compared to placebo as add-on on basal
An observational study on diabetic subjects [131] insulin; also in this trial, the degree of metabolic control
found that myocardial diastolic dysfunction (Em) was was superimposable in the two arms. In the SPREAD-
independently predicted by age (p Z 0.013), hypertension DIMCAD trial, the patients with DM2 and cardiovascular
(p Z 0.001), insulin (p Z 0.008), and, unexpectedly, disease treated with metformin showed a 46% reduction in
metformin (p Z 0.01) treatment. However, it has to be recurrent cardiovascular events (as composite endpoint)
pointed out that the subjects treated with metformin had when compared to the active comparator (glipizide) [136].
a higher BMI value; as a consequence, the weak, adverse These results stand in contrast with the neutral results of
effect of metformin on diastolic function might be due to the BARI2D trial performed in type 2 diabetic patients who
this confounding factor. In a more recent study [132], were eligible for coronary artery revascularization [137].
metformin therapy was an independent predictor of left This study, however, compared two therapeutic strategies,
ventricular dysfunction. Nonetheless, in the same study, insulin sensitizing (metformin and/or thiazolidinediones)
waist circumference and metformin were adversely versus insulin providing (SUDs and/or insulin) drugs,
associated with systolic dysfunction (OR 1.02, 95% CI therefore it does not provide direct information on the ef-
[1.01e1.04] and 1.57, 95% CI [1.01e2.43], respectively), fect of metformin per se.
confirming that in this setting metformin therapy might In 2010, Roussel et al. [138] demonstrated that data
rather represent a marker of visceral adiposity. Other from the observational Reduction of Atherothrombosis for
Metformin effects on the heart and the cardiovascular system 665

Continued Health Registry indicated that the use of met- mainly due to a decreased cardiovascular mortality, irre-
formin was associated with a significant 24% reduction in spective of gender, age, and etiology of HF. Such hypothesis
all-cause mortality after a 2-year follow-up. Meta-analyses requires to be challenged in an ad-hoc designed random-
of observational studies confirm that, compared to sulfo- ized clinical trial, the power of which could be increased
nylureas, metformin is related to a reduced risk of MI using mechanism-based biomarkers of metformin action
[139], and that metformin monotherapy can improve rather than parameters of diabetes control.
survival versus placebo or no treatment or SUDs [109].
Nevertheless, when randomized clinical trials have been
Concluding remarks
meta-analyzed, the effectiveness of metformin to reduce
cardiovascular events was not so evident, particularly in
Metformin in experimental studies exerts positive effects
the smaller and shorter studies [109,140].
on cardiac energetic metabolism, structure, and function
Concluding the evidence that metformin reduces the
through reduced oxidative stress and inflammation, and
risk of cardiovascular events though limited is rather
improved endothelial function. It reduces ROS production
consistent, particularly with regard to sulfonylureas. This is
by inhibiting the activity of protein kinase C-NAD(P)H
somewhat in keeping with the positive effects of the car-
oxidase pathway, promotes macrophage differentiation to
diovascular systems observed in experimental studies,
the anti-inflammatory functional phenotype, increases the
which also suggest that the effects are proportional to the
expression of antioxidant genes, and increases eNOS ac-
load of metabolic and/or hemodynamic stress. Accord-
tivity. In intact animals, metformin appears to attenuate
ingly, metformin could be particularly effective in specific
cardiac remodeling by reducing fibrosis, hypertrophy, and
subgroups of patients; for example in sedentary or obese
inflammation-induced damage, thereby preserving LV
patients with less-controlled blood pressure as suggested
morphology and systo-diastolic performance from meta-
by the impressive results of the UKPDS.
bolic, hemodynamic, and ischemic-induced damages. The
overall evidence emerging from both clinical trials and real
Heart failure world registry is in favor of a protective effect of metfor-
min with regard to both coronary events and progression
Evans et al. [141] demonstrated that patients with DM2 to HF. Given this potential, its efficacy, and its safety (also
and HF who were treated with metformin alone or in its low cost), metformin remains the central pillar of the
combination with SUDs were at significantly lower risk of therapy of diabetes. Therefore, a large clinical trial
all-cause mortality during a 1-year follow-up than those designed ad hoc to estimate with more precision the size
who were treated with SUDs alone. In addition, Aguilar of its effects on these cardiovascular outcomes would be
et al. [142] confirmed that metformin therapy was asso- very difficult, if not unethical; however, hypothesis-driven
ciated with a survival benefit over a 2-year follow-up. small trials on specific subgroups of patients focusing on
Importantly, this benefit was consistent across sub- robust surrogate endpoints might help in refining our
groups of diabetic patients with HF, and several comor- knowledge and would allow the full exploitation of the
bidities usually excluded from clinical trials, such as renal potential of this drug.
insufficiency, prior MI, and obesity. The study of Romero
et al. [143] demonstrated in more than 1500 patients with
References
new-onset DM and HF that initiation of metformin ther-
apy was associated not only with a decrease in all-cause
[1] Natali A, Ferrannini E. Effects of metformin and thiazolidine-
and cardiovascular mortality but also with a lower hos- diones on suppression of hepatic glucose production and stim-
pitalization rate for HF in a 2-year follow-up. In this ulation of glucose uptake in type 2 diabetes: a systematic review.
setting, a recent interesting study evaluating the effect of Diabetologia 2006;49:434e41.
[2] Eurich DT, Majumdar SR, McAlister FA, Tsuyuki RT, Johnson JA.
metformin in insulin-resistant adults with HF on exercise Improved clinical outcomes associated with metformin in pa-
showed that metformin increased the VE/VCO2 slope (the tients with diabetes and heart failure. Diabetes Care 2005;28:
ratio of minute ventilation to carbon dioxide production, 2345e51.
which in this population is a more sensitive index of the [3] Inzucchi SE, Masoudi FA, McGuire DK. Metformin in heart failure.
Diabetes Care 2007;30:e129.
cardio respiratory performance with regard to VO2max) [4] Masoudi FA, Inzucchi SE, Wang Y, Havranek EP, Foody JM,
[144]. In another small study in subjects with normal Krumholz HM. Thiazolidinediones, metformin, and outcomes in
cardiac function, metformin did not reduce VO2max but older patients with diabetes and heart failure: an observational
study. Circulation 2005;111:583e90.
increased the max workload [145]. This suggests a [5] Eurich DT, Weir DL, Majumdar SR, Tsuyuki RT, Johnson JA,
possible mechanism of action for metformin in reducing Tjosvold L, et al. Comparative safety and effectiveness of met-
HF-related outcomes. Importantly, in diabetic patients formin in patients with diabetes mellitus and heart failure: sys-
with HF, a recent meta-analysis of cohort studies tematic review of observational studies involving 34,000
patients. Circ Heart Fail 2013;6:395e402.
confirmed that metformin, with regard to other glucose- [6] Aschenbrenner DS. The FDA revises restrictions on metformin
lowering agents, is associated with a reduced mortality use in kidney impairment. Am J Nurs 2016;116:22e3.
and all-cause hospitalization across the different classes [7] Diamanti-Kandarakis E, Economou F, Palimeri S, Christakou C.
Metformin in polycystic ovary syndrome. Ann N. Y Acad Sci 2010;
of kidney dysfunction and EF [5].
1205:192e8.
In summary, the effects that metformin therapy appears [8] Pollak M. Metformin’s potential in oncology. Clin Adv Hematol
to have on the survival of patients with HF and DM2 are Oncol 2013;11:594e5.
666 L. Nesti, A. Natali

[9] Pollak M. Potential applications for biguanides in oncology. J Clin [31] Burcelin R. The antidiabetic gutsy role of metformin uncovered?
Invest 2013;123:3693e700. Gut 2014;63:706e7.
[10] Brownfoot FC, Hastie R, Hannan NJ, Cannon P, Tuohey L, Parry LJ, [32] Hundal RS, Krssak M, Dufour S, Laurent D, Lebon V,
et al. Metformin as a prevention and treatment for preeclampsia: Chandramouli V, et al. Mechanism by which metformin reduces
effects on soluble fms-like tyrosine kinase 1 and soluble endoglin glucose production in type 2 diabetes. Diabetes 2000;49:
secretion and endothelial dysfunction. Am J Obstet Gynecol 2016; 2063e9.
214. 356e1ee15. [33] Inzucchi SE, Maggs DG, Spollett GR, Page SL, Rife FS, Walton V,
[11] Nath N, Khan M, Paintlia MK, Singh I, Hoda MN, Giri S. Metformin et al. Efficacy and metabolic effects of metformin and troglita-
attenuated the autoimmune disease of the central nervous sys- zone in type II diabetes mellitus. N Engl J Med 1998;338:867e72.
tem in animal models of multiple sclerosis. J Immunol 2009;182: [34] Zhou ZY, Ren LW, Zhan P, Yang HY, Chai DD, Yu ZW. Metformin
8005e14. exerts glucose-lowering action in high-fat fed mice via attenu-
[12] Martin-Montalvo A, Mercken EM, Mitchell SJ, Palacios HH, ating endotoxemia and enhancing insulin signaling. Acta Phar-
Mote PL, Scheibye-Knudsen M, et al. Metformin improves macol Sin 2016;37:1063e75.
healthspan and lifespan in mice. Nat Commun 2013;4:2192. [35] Cani PD, Amar J, Iglesias MA, Poggi M, Knauf C, Bastelica D, et al.
[13] Holman RR, Paul SK, Bethel MA, Matthews DR, Neil HA. 10-year Metabolic endotoxemia initiates obesity and insulin resistance.
follow-up of intensive glucose control in type 2 diabetes. N Diabetes 2007;56:1761e72.
Engl J Med 2008;359:1577e89. [36] Shin NR, Lee JC, Lee HY, Kim MS, Whon TW, Lee MS, et al. An
[14] Zhou G, Myers R, Li Y, Chen Y, Shen X, Fenyk-Melody J, et al. Role increase in the Akkermansia spp. population induced by met-
of AMP-activated protein kinase in mechanism of metformin formin treatment improves glucose homeostasis in diet-induced
action. J Clin Invest 2001;108:1167e74. obese mice. Gut 2014;63:727e35.
[15] Hawley SA, Gadalla AE, Olsen GS, Hardie DG. The antidiabetic [37] Pandey A, Kumar VL. Protective effect of metformin against acute
drug metformin activates the AMP-activated protein kinase inflammation and oxidative stress in rat. Drug Dev Res 2016;77:
cascade via an adenine nucleotide-independent mechanism. 278e84.
Diabetes 2002;51:2420e5. [38] Hattori Y, Hattori K, Hayashi T. Pleiotropic benefits of metformin:
[16] Madiraju AK, Erion DM, Rahimi Y, Zhang XM, Braddock DT, macrophage targeting its anti-inflammatory mechanisms. Dia-
Albright RA, et al. Metformin suppresses gluconeogenesis by betes 2015;64:1907e9.
inhibiting mitochondrial glycerophosphate dehydrogenase. Na- [39] Kim J, Kwak HJ, Cha JY, Jeong YS, Rhee SD, Kim KR, et al. Met-
ture 2014;510:542e6. formin suppresses lipopolysaccharide (LPS)-induced inflamma-
[17] Coughlan KA, Valentine RJ, Ruderman NB, Saha AK. AMPK acti- tory response in murine macrophages via activating
vation: a therapeutic target for type 2 diabetes? Diabetes Metab transcription factor-3 (ATF-3) induction. J Biol Chem 2014;289:
Syndr Obes 2014;7:241e53. 23246e55.
[18] Shaw RJ, Lamia KA, Vasquez D, Koo SH, Bardeesy N, Depinho RA, [40] Iida KT, Kawakami Y, Suzuki M, Shimano H, Toyoshima H, Sone H,
et al. The kinase LKB1 mediates glucose homeostasis in liver and et al. Effect of thiazolidinediones and metformin on LDL oxida-
therapeutic effects of metformin. Science 2005;310:1642e6. tion and aortic endothelium relaxation in diabetic GK rats. Am J
[19] Ouyang J, Parakhia RA, Ochs RS. Metformin activates AMP kinase Physiol Endocrinol Metab 2003;284:E1125e30.
through inhibition of AMP deaminase. J Biol Chem 2011;286:1e11. [41] Rabbani N, Chittari MV, Bodmer CW, Zehnder D, Ceriello A,
[20] Owen MR, Doran E, Halestrap AP. Evidence that metformin exerts Thornalley PJ. Increased glycation and oxidative damage to
its anti-diabetic effects through inhibition of complex 1 of the apolipoprotein B100 of LDL cholesterol in patients with type 2
mitochondrial respiratory chain. Biochem J 2000;348:607e14. diabetes and effect of metformin. Diabetes 2010;59:1038e45.
[21] Mihaylova MM, Shaw RJ. The AMPK signalling pathway co- [42] Buldak L, Labuzek K, Buldak RJ, Kozlowski M, Machnik G, Liber S,
ordinates cell growth, autophagy and metabolism. Nat Cell Biol et al. Metformin affects macrophages’ phenotype and improves
2011;13:1016e23. the activity of glutathione peroxidase, superoxide dismutase,
[22] Sakamoto K, Holman GD. Emerging role for AS160/TBC1D4 and catalase and decreases malondialdehyde concentration in a
TBC1D1 in the regulation of GLUT4 traffic. Am J Physiol Endo- partially AMPK-independent manner in LPS-stimulated human
crinol Metab 2008;295:E29e37. monocytes/macrophages. Pharmacol Rep 2014;66:418e29.
[23] Li Y, Xu S, Mihaylova MM, Zheng B, Hou X, Jiang B, et al. AMPK [43] Buldak L, Labuzek K, Buldak RJ, Machnik G, Boldys A, Basiak M,
phosphorylates and inhibits SREBP activity to attenuate hepatic et al. Metformin reduces the expression of NADPH oxidase and
steatosis and atherosclerosis in diet-induced insulin-resistant increases the expression of antioxidative enzymes in human
mice. Cell Metab 2011;13:376e88. monocytes/macrophages cultured in vitro. Exp Ther Med 2016;
[24] Canto C, Auwerx J. AMP-activated protein kinase and its down- 11:1095e103.
stream transcriptional pathways. Cell Mol Life Sci 2010;67: [44] Vasamsetti SB, Karnewar S, Kanugula AK, Thatipalli AR,
3407e23. Kumar JM, Kotamraju S. Metformin inhibits monocyte-to-
[25] Camporez JP, Kanda S, Petersen MC, Jornayvaz FR, Samuel VT, macrophage differentiation via AMPK-mediated inhibition of
Bhanot S, et al. ApoA5 knockdown improves whole-body insulin STAT3 activation: potential role in atherosclerosis. Diabetes 2015;
sensitivity in high-fat-fed mice by reducing ectopic lipid content. 64:2028e41.
J Lipid Res 2015;56:526e36. [45] Satapati S, Kucejova B, Duarte JA, Fletcher JA, Reynolds L, Sunny NE,
[26] Fullerton MD, Galic S, Marcinko K, Sikkema S, Pulinilkunnil T, et al. Mitochondrial metabolism mediates oxidative stress and
Chen ZP, et al. Single phosphorylation sites in Acc1 and Acc2 inflammation in fatty liver. J Clin Invest 2015;125:4447e62.
regulate lipid homeostasis and the insulin-sensitizing effects of [46] Mahrouf M, Ouslimani N, Peynet J, Djelidi R, Couturier M,
metformin. Nat Med 2013;19:1649e54. Therond P, et al. Metformin reduces angiotensin-mediated
[27] Wang PY, Caspi L, Lam CK, Chari M, Li X, Light PE, et al. Upper intracellular production of reactive oxygen species in endothe-
intestinal lipids trigger a gut-brain-liver axis to regulate glucose lial cells through the inhibition of protein kinase C. Biochem
production. Nature 2008;452:1012e6. Pharmacol 2006;72:176e83.
[28] Duca FA, Cote CD, Rasmussen BA, Zadeh-Tahmasebi M, Rutter GA, [47] Batandier C, Guigas B, Detaille D, El-Mir MY, Fontaine E,
Filippi BM, et al. Metformin activates a duodenal Ampk- Rigoulet M, et al. The ROS production induced by a reverse-
dependent pathway to lower hepatic glucose production in electron flux at respiratory-chain complex 1 is hampered by
rats. Nat Med 2015;21:506e11. metformin. J Bioenerg Biomembr 2006;38:33e42.
[29] Foretz M, Hebrard S, Leclerc J, Zarrinpashneh E, Soty M, [48] Kelly B, Tannahill GM, Murphy MP, O’Neill LA. Metformin inhibits
Mithieux G, et al. Metformin inhibits hepatic gluconeogenesis in the production of reactive oxygen species from NADH: ubiqui-
mice independently of the LKB1/AMPK pathway via a decrease in none oxidoreductase to limit induction of Interleukin-1beta (IL-
hepatic energy state. J Clin Invest 2010;120:2355e69. 1beta) and boosts Interleukin-10 (IL-10) in lipopolysaccharide
[30] Stephenne X, Foretz M, Taleux N, van der Zon GC, Sokal E, Hue L, (LPS)-activated macrophages. J Biol Chem 2015;290:20348e59.
et al. Metformin activates AMP-activated protein kinase in pri- [49] Cameron AR, Morrison VL, Levin D, Mohan M, Forteath C, Beall C,
mary human hepatocytes by decreasing cellular energy status. et al. Anti-inflammatory effects of metformin irrespective of
Diabetologia 2011;54:3101e10. diabetes status. Circ Res 2016;119:652e65.
Metformin effects on the heart and the cardiovascular system 667

[50] Onken B, Driscoll M. Metformin induces a dietary restriction-like [68] Lee JH, Kim JH, Kim JS, Chang JW, Kim SB, Park JS, et al. AMP-
state and the oxidative stress response to extend C. elegans activated protein kinase inhibits TGF-beta-, angiotensin II-,
Healthspan via AMPK, LKB1, and SKN-1. PLoS One 2010;5:e8758. aldosterone-, high glucose-, and albumin-induced epithelial-
[51] Chin JT, Troke JJ, Kimura N, Itoh S, Wang X, Palmer OP, et al. A mesenchymal transition. Am J Physiol Ren Physiol 2013;304:
novel cardioprotective agent in cardiac transplantation: metfor- F686e97.
min activation of AMP-activated protein kinase decreases acute [69] Hernandez JS, Barreto-Torres G, Kuznetsov AV, Khuchua Z,
ischemia-reperfusion injury and chronic rejection. Yale J Biol Javadov S. Crosstalk between AMPK activation and angiotensin
Med 2011;84:423e32. II-induced hypertrophy in cardiomyocytes: the role of mito-
[52] Batchuluun B, Inoguchi T, Sonoda N, Sasaki S, Inoue T, Fujimura Y, chondria. J Cell Mol Med 2014;18:709e20.
et al. Metformin and liraglutide ameliorate high glucose-induced [70] Forouzandeh F, Salazar G, Patrushev N, Xiong S, Hilenski L, Fei B,
oxidative stress via inhibition of PKC-NAD(P)H oxidase pathway et al. Metformin beyond diabetes: pleiotropic benefits of met-
in human aortic endothelial cells. Atherosclerosis 2014;232: formin in attenuation of atherosclerosis. J Am Heart Assoc 2014;
156e64. 3:e001202.
[53] Bhatt MP, Lim YC, Kim YM, Ha KS. C-peptide activates AMPKal- [71] Petersen JS, DiBona GF. Acute sympathoinhibitory actions of
pha and prevents ROS-mediated mitochondrial fission and metformin in spontaneously hypertensive rats. Hypertension
endothelial apoptosis in diabetes. Diabetes 2013;62:3851e62. 1996;27:619e25.
[54] Pyla R, Osman I, Pichavaram P, Hansen P, Segar L. Metformin [72] Petersen J. Intracerebroventricular metformin attenuates salt-
exaggerates phenylephrine-induced AMPK phosphorylation in- induced hypertension in spontaneously hypertensive rats. Am J
dependent of CaMKKbeta and attenuates contractile response in Hypertens 2001;14:1116e22.
endothelium-denuded rat aorta. Biochem Pharmacol 2014;92: [73] Muntzel MS, Hamidou I, Barrett S. Metformin attenuates salt-
266e79. induced hypertension in spontaneously hypertensive rats. Hy-
[55] Shang F, Zhang J, Li Z, Zhang J, Yin Y, Wang Y, et al. Cardiovascular pertension 1999;33:1135e40.
protective effect of metformin and telmisartan: reduction of [74] Giugliano D, De Rosa N, Di Maro G, Marfella R, Acampora R,
PARP1 activity via the AMPK-PARP1 cascade. PLoS One 2016;11: Buoninconti R, et al. Metformin improves glucose, lipid meta-
e0151845. bolism, and reduces blood pressure in hypertensive, obese
[56] Davis BJ, Xie Z, Viollet B, Zou MH. Activation of the AMP-activated women. Diabetes Care 1993;16:1387e90.
kinase by antidiabetes drug metformin stimulates nitric oxide [75] Landin K, Tengborn L, Smith U. Treating insulin resistance in
synthesis in vivo by promoting the association of heat shock hypertension with metformin reduces both blood-pressure and
protein 90 and endothelial nitric oxide synthase. Diabetes 2006; metabolic risk-factors. J Intern Med 1991;229:181e7.
55:496e505. [76] Manzella D, Grella R, Esposito K, Giugliano D, Barbagallo M,
[57] Massion PB, Feron O, Dessy C, Balligand JL. Nitric oxide and Paolisso G. Blood pressure and cardiac autonomic nervous sys-
cardiac function: ten years after, and continuing. Circ Res 2003; tem in obese type 2 diabetic patients: effect of metformin
93:388e98. administration. Am J Hypertens 2004;17:223e7.
[58] Lai YC, Tabima DM, Dube JJ, Hughan KS, Vanderpool RR, [77] Nagi DK, Yudkin JS. Effects of metformin on insulin resistance,
Goncharov DA, et al. SIRT3-AMP-Activated protein kinase acti- risk factors for cardiovascular disease, and plasminogen activator
vation by nitrite and metformin improves hyperglycemia and inhibitor in NIDDM subjects. A study of two ethnic groups. Dia-
normalizes pulmonary hypertension associated with heart fail- betes Care 1993;16:621e9.
ure with preserved ejection fraction. Circulation 2016;133: [78] Uehara MH, Kohlmann NE, Zanella MT, Ferreira SR. Metabolic
717e31. and haemodynamic effects of metformin in patients with type 2
[59] Dean A, Nilsen M, Loughlin L, Salt IP, MacLean MR. Metformin diabetes mellitus and hypertension. Diabetes Obes Metab 2001;
reverses development of pulmonary hypertension via aromatase 3:319e25.
inhibition. Hypertension 2016;68:446e54. [79] Wulffele MG, Kooy A, Lehert P, Bets D, Donker AJ, Stehouwer CD.
[60] Song Y, Wu Y, Su X, Zhu Y, Liu L, Pan Y, et al. Activation of AMPK Does metformin decrease blood pressure in patients with Type 2
inhibits PDGF-induced pulmonary arterial smooth muscle cells diabetes intensively treated with insulin? Diabet Med 2005;22:
proliferation and its potential mechanisms. Pharmacol Res 2016; 907e13.
107:117e24. [80] Charles MA, Eschwege E, Grandmottet P, Isnard F, Cohen JM,
[61] Haffner S, Temprosa M, Crandall J, Fowler S, Goldberg R, Bensoussan JL, et al. Treatment with metformin of non-diabetic
Horton E, et al. Intensive lifestyle intervention or metformin on men with hypertension, hypertriglyceridaemia and central fat
inflammation and coagulation in participants with impaired distribution: the BIGPRO 1.2 trial. Diabetes Metab Res Rev 2000;
glucose tolerance. Diabetes 2005;54:1566e72. 16:2e7.
[62] Morin-Papunen L, Rautio K, Ruokonen A, Hedberg P, Puukka M, [81] Dorella M, Giusto M, Da Tos V, Campagnolo M, Palatini P, Rossi G,
Tapanainen JS. Metformin reduces serum C-reactive protein et al. Improvement of insulin sensitivity by metformin treatment
levels in women with polycystic ovary syndrome. J Clin Endo- does not lower blood pressure of nonobese insulin-resistant
crinol Metab 2003;88:4649e54. hypertensive patients with normal glucose tolerance. J Clin
[63] Sobel BE, Hardison RM, Genuth S, Brooks MM, McBane 3rd RD, Endocrinol Metab 1996;81:1568e74.
Schneider DJ, et al. Profibrinolytic, antithrombotic, and antiin- [82] Gudbjornsdottir S, Friberg P, Elam M, Attvall S, Lonnroth P,
flammatory effects of an insulin-sensitizing strategy in patients Wallin BG. The effect of metformin and insulin on sympathetic
in the Bypass Angioplasty Revascularization Investigation 2 nerve activity, norepinephrine spillover and blood pressure in
Diabetes (BARI 2D) trial. Circulation 2011;124:695e703. obese, insulin resistant, normoglycemic, hypertensive men.
[64] de Jager J, Kooy A, Schalkwijk C, van der Kolk J, Lehert P, Bets D, Blood Press 1994;3:394e403.
et al. Long-term effects of metformin on endothelial function in [83] Snorgaard O, Kober L, Carlsen J. The effect of metformin on blood
type 2 diabetes: a randomized controlled trial. J Intern Med 2014; pressure and metabolism in nondiabetic hypertensive patients. J
275:59e70. Intern Med 1997;242:407e12.
[65] Jadhav S, Ferrell W, Greer IA, Petrie JR, Cobbe SM, Sattar N. Effects [84] Malin SK, Nightingale J, Choi SE, Chipkin SR, Braun B. Metformin
of metformin on microvascular function and exercise tolerance in modifies the exercise training effects on risk factors for cardio-
women with angina and normal coronary arteries: a randomized, vascular disease in impaired glucose tolerant adults. Obes (Silver
double-blind, placebo-controlled study. J Am Coll Cardiol 2006; Spring) 2013;21:93e100.
48:956e63. [85] Orchard TJ, Temprosa M, Goldberg R, Haffner S, Ratner R,
[66] Mather KJ, Verma S, Anderson TJ. Improved endothelial function Marcovina S, et al. The effect of metformin and intensive lifestyle
with metformin in type 2 diabetes mellitus. J Am Coll Cardiol intervention on the metabolic syndrome: the Diabetes Preven-
2001;37:1344e50. tion Program randomized trial. Ann Intern Med 2005;142:611e9.
[67] Natali A, Baldeweg S, Toschi E, Capaldo B, Barbaro D, [86] Ratner R, Goldberg R, Haffner S, Marcovina S, Orchard T, Fowler S,
Gastaldelli A, et al. Vascular effects of improving metabolic et al. Impact of intensive lifestyle and metformin therapy on
control with metformin or rosiglitazone in type 2 diabetes. cardiovascular disease risk factors in the diabetes prevention
Diabetes Care 2004;27:1349e57. program. Diabetes Care 2005;28:888e94.
668 L. Nesti, A. Natali

[87] Zhou L, Liu H, Wen X, Peng Y, Tian Y, Zhao L. Effects of metformin cardioprotection against infarction: not just a glucose lowering
on blood pressure in nondiabetic patients: a meta-analysis of phenomenon. Cardiovasc Drugs Ther 2013;27:5e16.
randomized controlled trials. J Hypertens 2017;35:18e26. [106] Elmadhun NY, Sabe AA, Lassaletta AD, Chu LM, Sellke FW. Met-
[88] Costa EC, Goncalves AA, Areas MA, Morgabel RG. Effects of formin mitigates apoptosis in ischemic myocardium. J Surg Res
metformin on QT and QTc interval dispersion of diabetic rats. Arq 2014;192:50e8.
Bras Cardiol 2008;90:232e8. [107] Bhamra GS, Hausenloy DJ, Davidson SM, Carr RD, Paiva M,
[89] Soraya H, Khorrami A, Garjani A, Maleki-Dizaji N, Garjani A. Wynne AM, et al. Metformin protects the ischemic heart by the
Acute treatment with metformin improves cardiac function Akt-mediated inhibition of mitochondrial permeability transition
following isoproterenol induced myocardial infarction in rats. pore opening. Basic Res Cardiol 2008;103:274e84.
Pharmacol Rep 2012;64:1476e84. [108] Mellbin LG, Malmberg K, Norhammar A, Wedel H, Ryden L,
[90] Najeed SA, Khan IA, Molnar J, Somberg JC. Differential effect of Investigators D. Prognostic implications of glucose-lowering
glyburide (glibenclamide) and metformin on QT dispersion: a treatment in patients with acute myocardial infarction and dia-
potential adenosine triphosphate sensitive Kþ channel effect. betes: experiences from an extended follow-up of the Diabetes
Am J Cardiol 2002;90:1103e6. Mellitus Insulin-Glucose Infusion in Acute Myocardial Infarction
[91] Zarrinpashneh E, Carjaval K, Beauloye C, Ginion A, Mateo P, (DIGAMI) 2 Study. Diabetologia 2011;54:1308e17.
Pouleur AC, et al. Role of the alpha2-isoform of AMP-activated [109] Lamanna C, Monami M, Marchionni N, Mannucci E. Effect of
protein kinase in the metabolic response of the heart to no- metformin on cardiovascular events and mortality: a meta-
flow ischemia. Am J Physiol Heart Circ Physiol 2006;291: analysis of randomized clinical trials. Diabetes Obes Metab
H2875e83. 2011;13:221e8.
[92] Bertrand L, Ginion A, Beauloye C, Hebert AD, Guigas B, Hue L, [110] Zhao JL, Fan CM, Yang YJ, You SJ, Gao X, Zhou Q, et al. Chronic
et al. AMPK activation restores the stimulation of glucose uptake pretreatment of metformin is associated with the reduction of
in an in vitro model of insulin-resistant cardiomyocytes via the the no-reflow phenomenon in patients with diabetes mellitus
activation of protein kinase B. Am J Physiol Heart Circ Physiol after primary angioplasty for acute myocardial infarction. Car-
2006;291:H239e50. diovasc Ther 2013;31:60e4.
[93] Saeedi R, Parsons HL, Wambolt RB, Paulson K, Sharma V, Dyck JR, [111] Lexis CP, Wieringa WG, Hiemstra B, van Deursen VM, Lipsic E, van
et al. Metabolic actions of metformin in the heart can occur by der Harst P, et al. Chronic metformin treatment is associated with
AMPK-independent mechanisms. Am J Physiol Heart Circ Physiol reduced myocardial infarct size in diabetic patients with ST-
2008;294:H2497e506. segment elevation myocardial infarction. Cardiovasc Drugs Ther
[94] Pascual F, Coleman RA. Fuel availability and fate in cardiac 2014;28:163e71.
metabolism: a tale of two substrates. Biochim Biophys Acta 1860; [112] Basnet S, Kozikowski A, Makaryus AN, Pekmezaris R, Zeltser R,
2016:1425e33. Akerman M, et al. Metformin and myocardial injury in patients
[95] Shoghi KI, Finck BN, Schechtman KB, Sharp T, Herrero P, with diabetes and ST-segment elevation myocardial infarction: a
Gropler RJ, et al. In vivo metabolic phenotyping of myocardial propensity score matched analysis. J Am Heart Assoc 2015;4:
substrate metabolism in rodents: differential efficacy of metfor- e002314.
min and rosiglitazone monotherapy. Circ Cardiovasc Imaging [113] Lexis CP, van der Horst IC, Lipsic E, Wieringa WG, de Boer RA, van
2009;2:373e81. den Heuvel AF, et al. Effect of metformin on left ventricular
[96] Hallsten K, Virtanen KA, Lonnqvist F, Janatuinen T, Turiceanu M, function after acute myocardial infarction in patients without
Ronnemaa T, et al. Enhancement of insulin-stimulated myocar- diabetes: the GIPS-III randomized clinical trial. JAMA 2014;311:
dial glucose uptake in patients with Type 2 diabetes treated with 1526e35.
rosiglitazone. Diabet Med 2004;21:1280e7. [114] Preiss D, Lloyd SM, Ford I, McMurray JJ, Holman RR, Welsh P, et al.
[97] van der Meer RW, Rijzewijk LJ, de Jong HW, Lamb HJ, Metformin for non-diabetic patients with coronary heart disease
Lubberink M, Romijn JA, et al. Pioglitazone improves cardiac (the CAMERA study): a randomised controlled trial. Lancet Dia-
function and alters myocardial substrate metabolism without betes Endocrinol 2014;2:116e24.
affecting cardiac triglyceride accumulation and high-energy [115] El Messaoudi S, Nederlof R, Zuurbier CJ, van Swieten HA,
phosphate metabolism in patients with well-controlled type 2 Pickkers P, Noyez L, et al. Effect of metformin pretreatment on
diabetes mellitus. Circulation 2009;119:2069e77. myocardial injury during coronary artery bypass surgery in pa-
[98] Lyons MR, Peterson LR, McGill JB, Herrero P, Coggan AR, tients without diabetes (MetCAB): a double-blind, randomised
Saeed IM, et al. Impact of sex on the heart’s metabolic and controlled trial. Lancet Diabetes Endocrinol 2015;3:615e23.
functional responses to diabetic therapies. Am J Physiol Heart [116] Li J, Xu JP, Zhao XZ, Sun XJ, Xu ZW, Song SJ. Protective effect of
Circ Physiol 2013;305:H1584e91. metformin on myocardial injury in metabolic syndrome patients
[99] Legtenberg RJ, Houston RJ, Oeseburg B, Smits P. Metformin im- following percutaneous coronary intervention. Cardiology 2014;
proves cardiac functional recovery after ischemia in rats. Horm 127:133e9.
Metab Res 2002;34:182e5. [117] Zhang CX, Pan SN, Meng RS, Peng CQ, Xiong ZJ, Chen BL, et al.
[100] Calvert JW, Gundewar S, Jha S, Greer JJ, Bestermann WH, Tian R, Metformin attenuates ventricular hypertrophy by activating the
et al. Acute metformin therapy confers cardioprotection against AMP-activated protein kinase-endothelial nitric oxide synthase
myocardial infarction via AMPK-eNOS-mediated signaling. Dia- pathway in rats. Clin Exp Pharmacol Physiol 2011;38:55e62.
betes 2008;57:696e705. [118] Xiao H, Ma X, Feng W, Fu Y, Lu Z, Xu M, et al. Metformin atten-
[101] Yin M, van der Horst IC, van Melle JP, Qian C, van Gilst WH, uates cardiac fibrosis by inhibiting the TGFbeta1-Smad3 signal-
Sillje HH, et al. Metformin improves cardiac function in a ling pathway. Cardiovasc Res 2010;87:504e13.
nondiabetic rat model of post-MI heart failure. Am J Physiol [119] Wang XF, Zhang JY, Li L, Zhao XY. Beneficial effects of metformin
Heart Circ Physiol 2011;301:H459e68. on primary cardiomyocytes via activation of adenosine
[102] Gundewar S, Calvert JW, Jha S, Toedt-Pingel I, Ji SY, Nunez D, et al. monophosphate-activated protein kinase. Chin Med J Engl 2011;
Activation of AMP-activated protein kinase by metformin im- 124:1876e84.
proves left ventricular function and survival in heart failure. Circ [120] Sasaki H, Asanuma H, Fujita M, Takahama H, Wakeno M, Ito S, et al.
Res 2009;104:403e11. Metformin prevents progression of heart failure in dogs: role of
[103] Solskov L, Lofgren B, Kristiansen SB, Jessen N, Pold R, Nielsen TT, AMP-activated protein kinase. Circulation 2009;119:2568e77.
et al. Metformin induces cardioprotection against ischaemia/r- [121] Xu X, Lu Z, Fassett J, Zhang P, Hu X, Liu X, et al. Metformin pro-
eperfusion injury in the rat heart 24 hours after administration. tects against systolic overload-induced heart failure independent
Basic Clin Pharmacol Toxicol 2008;103:82e7. of AMP-activated protein kinase alpha2. Hypertension 2014;63:
[104] Paiva M, Riksen NP, Davidson SM, Hausenloy DJ, Monteiro P, 723e8.
Goncalves L, et al. Metformin prevents myocardial reperfusion [122] Wang XF, Zhang JY, Li L, Zhao XY, Tao HL, Zhang L. Metformin
injury by activating the adenosine receptor. J Cardiovasc Phar- improves cardiac function in rats via activation of AMP-activated
macol 2009;53:373e8. protein kinase. Clin Exp Pharmacol Physiol 2011;38:94e101.
[105] Whittington HJ, Hall AR, McLaughlin CP, Hausenloy DJ, [123] Noppe G, Dufeys C, Buchlin P, Marquet N, Castanares-Zapatero D,
Yellon DM, Mocanu MM. Chronic metformin associated Balteau M, et al. Reduced scar maturation and contractility lead
Metformin effects on the heart and the cardiovascular system 669

to exaggerated left ventricular dilation after myocardial infarc- [135] Kooy A, de Jager J, Lehert P, Bets D, Wulffele MG, Donker AJ, et al.
tion in mice lacking AMPKalpha1. J Mol Cell Cardiol 2014;74: Long-term effects of metformin on metabolism and microvas-
32e43. cular and macrovascular disease in patients with type 2 diabetes
[124] Cittadini A, Napoli R, Monti MG, Rea D, Longobardi S, Netti PA, mellitus. Arch Intern Med 2009;169:616e25.
et al. Metformin prevents the development of chronic heart [136] Hong J, Zhang Y, Lai S, Lv A, Su Q, Dong Y, et al. Effects of met-
failure in the SHHF rat model. Diabetes 2012;61:944e53. formin versus glipizide on cardiovascular outcomes in patients
[125] Xie Z, Lau K, Eby B, Lozano P, He C, Pennington B, et al. with type 2 diabetes and coronary artery disease. Diabetes Care
Improvement of cardiac functions by chronic metformin treat- 2013;36:1304e11.
ment is associated with enhanced cardiac autophagy in diabetic [137] Group BDS, Frye RL, August P, Brooks MM, Hardison RM,
OVE26 mice. Diabetes 2011;60:1770e8. Kelsey SF, et al. A randomized trial of therapies for type 2 dia-
[126] Mohan M, McSwiggan S, Baig F, Rutherford L, Lang CC. Metformin betes and coronary artery disease. N Engl J Med 2009;360:
and its effects on myocardial dimension and left ventricular hy- 2503e15.
pertrophy in normotensive patients with coronary heart disease [138] Roussel R, Travert F, Pasquet B, Wilson PW, Smith Jr SC, Goto S,
(the MET-REMODEL study): rationale and design of the MET- et al. Metformin use and mortality among patients with diabetes
REMODEL study. Cardiovasc Ther 2015;33:1e8. and atherothrombosis. Arch Intern Med 2010;170:1892e9.
[127] Benes J, Kazdova L, Drahota Z, Houstek J, Medrikova D, Kopecky J, [139] Pladevall M, Riera-Guardia N, Margulis AV, Varas-Lorenzo C,
et al. Effect of metformin therapy on cardiac function and sur- Calingaert B, Perez-Gutthann S. Cardiovascular risk associated
vival in a volume-overload model of heart failure in rats. Clin Sci with the use of glitazones, metformin and sufonylureas: meta-
(Lond) 2011;121:29e41. analysis of published observational studies. BMC Cardiovasc
[128] Verma S, McNeill JH. Metformin improves cardiac function in Disord 2016;16:14.
isolated streptozotocin-diabetic rat hearts. Am J Physiol 1994; [140] Boussageon R, Supper I, Bejan-Angoulvant T, Kellou N,
266:H714e9. Cucherat M, Boissel JP, et al. Reappraisal of metformin efficacy in
[129] Sen S, Kundu BK, Wu HC, Hashmi SS, Guthrie P, Locke LW, et al. the treatment of type 2 diabetes: a meta-analysis of randomised
Glucose regulation of load-induced mTOR signaling and ER stress controlled trials. PLoS Med 2012;9:e1001204.
in mammalian heart. J Am Heart Assoc 2013;2:e004796. [141] Evans JM, Doney AS, AlZadjali MA, Ogston SA, Petrie JR, Morris AD,
[130] Jyothirmayi GN, Soni BJ, Masurekar M, Lyons M, Regan TJ. Effects of et al. Effect of Metformin on mortality in patients with heart failure
metformin on collagen glycation and diastolic dysfunction in dia- and type 2 diabetes mellitus. Am J Cardiol 2010;106:1006e10.
betic myocardium. J Cardiovasc Pharmacol Ther 1998;3:319e26. [142] Aguilar D, Chan W, Bozkurt B, Ramasubbu K, Deswal A. Metfor-
[131] Fang ZY, Schull-Meade R, Downey M, Prins J, Marwick TH. De- min use and mortality in ambulatory patients with diabetes and
terminants of subclinical diabetic heart disease. Diabetologia heart failure. Circ Heart Fail 2011;4:53e8.
2005;48:394e402. [143] Romero SP, Andrey JL, Garcia-Egido A, Escobar MA, Perez V,
[132] Giorda CB, Cioffi G, de Simone G, Di Lenarda A, Faggiano P, Corzo R, et al. Metformin therapy and prognosis of patients
Latini R, et al. Predictors of early-stage left ventricular dysfunc- with heart failure and new-onset diabetes mellitus. A
tion in type 2 diabetes: results of DYDA study. Eur J Cardiovasc propensity-matched study in the community. Int J Cardiol
Prev Rehabil 2011;18:415e23. 2013;166:404e12.
[133] Leung M, Wong VW, Hudson M, Leung DY. Impact of improved [144] Wong AK, Symon R, AlZadjali MA, Ang DS, Ogston S, Choy A, et al.
glycemic control on cardiac function in type 2 diabetes mellitus. The effect of metformin on insulin resistance and exercise pa-
Circ Cardiovasc Imaging 2016;9:e003643. rameters in patients with heart failure. Eur J Heart Fail 2012;14:
[134] Effect of intensive blood-glucose control with metformin on 1303e10.
complications in overweight patients with type 2 diabetes [145] Cadeddu C, Nocco S, Deidda M, Cadeddu F, Bina A, Demuru P,
(UKPDS 34). UK Prospective Diabetes Study (UKPDS) Group. et al. Relationship between high values of HOMA-IR and car-
Lancet 1998;352:854e65. diovascular response to metformin. Int J Cardiol 2013;167:282.

Potrebbero piacerti anche