ArticlePDF AvailableLiterature Review

The protective effects of 17-β estradiol and SIRT1 against cardiac hypertrophy: a review

Authors:

Abstract and Figures

One of the major causes of morbidity and mortality worldwide is cardiac hypertrophy (CH), which leads to heart failure. Sex differences in CH can be caused by sex hormones or their receptors. The incidence of CH increases in postmenopausal women due to the decrease in female sex hormone 17-β estradiol (E2) during menopause. E2 and its receptors inhibit CH in humans and animal models. Silent information regulator 1 (SIRT1) is a NAD+-dependent HDAC (histone deacetylase) and plays a major role in biological processes, such as inflammation, apoptosis, and oxidative stress responses. Probably SIRT1 because of these effects, is one of the main suppressors of CH and has a cardioprotective effect. On the other hand, estrogen and its agonists are highly efficient in modulating SIRT1 expression. In the present study, we review the protective effects of E2 and SIRT1 against CH.
This content is subject to copyright. Terms and conditions apply.
Vol.:(0123456789)
1 3
Heart Failure Reviews
https://doi.org/10.1007/s10741-021-10171-0
The protective effects of17‑β estradiol andSIRT1 againstcardiac
hypertrophy: areview
ZahraHajializadeh1· MohammadKhaksari2
Accepted: 7 September 2021
© The Author(s), under exclusive licence to Springer Science+Business Media, LLC, part of Springer Nature 2021
Abstract
One of the major causes of morbidity and mortality worldwide is cardiac hypertrophy (CH), which leads to heart failure.
Sex differences in CH can be caused by sex hormones or their receptors. The incidence of CH increases in postmenopausal
women due to the decrease in female sex hormone 17-β estradiol (E2) during menopause. E2 and its receptors inhibit CH
in humans and animal models. Silent information regulator 1 (SIRT1) is a NAD+-dependent HDAC (histone deacetylase)
and plays a major role in biological processes, such as inflammation, apoptosis, and oxidative stress responses. Probably
SIRT1 because of these effects, is one of the main suppressors of CH and has a cardioprotective effect. On the other hand,
estrogen and its agonists are highly efficient in modulating SIRT1 expression. In the present study, we review the protective
effects of E2 and SIRT1 against CH.
Keywords 17-β estradiol· SIRT1· Cardiac hypertrophy· Estrogen receptors
Introduction
One of the major causes of morbidity and mortality world-
wide is cardiovascular disease [1]. There are 17.3 million
deaths each year and it is predicted to reach more than 23.6
million by 2030 [2]. Almost all types of heart failure are
associated with cardiac hypertrophy (CH) [3]. Systolic or
diastolic wall stress increases chronic physiological process
in cardiac muscle mass characterizing CH. This usually hap-
pens in response to sustained exercise, during pregnancy,
and during development [4]. Moreover, a number of patho-
logical conditions, such as valvular disease, hypertension,
cardiomyopathy, and myocardial infarction may cause it [5].
CH at the cellular level is determined by increasing cell size,
reactivation of fetal gene program, and protein synthesis,
eventually leading to heart failure [6].
Types ofcardiac hypertrophy
Physiological hypertrophy can be determined by the nor-
mal organization of cardiac structure along with natural or
increased contractile function [7]. Physiological hypertro-
phy is triggered by pregnancy, exercise (athlete's heart), or
growth factor stimulation [8]. Physiological hypertrophy is
identified by normal or increased cardiac function, normal
survival, and absence of pathological features like apopto-
sis and fibrosis [9]. Pathological hypertrophy is determined
by increased diastolic function and decreased systolic func-
tion, which often leads to heart failure and is associated with
increased cardiomyocyte death [4]. Pathological hypertrophy
is caused by various stimuli such as obesity, hypertension,
stenosis, or heart valve failure [4]. Early events that cause
CH are humoral stimulation and mechanical stress, which
are associated with cellular responses modulation, including
protein synthesis, sarcomere assembly, gene expression, cell
metabolism [1012]. Physiological hypertrophy differs from
pathological hypertrophy in terms of signaling pathways that
control these processes. Research studies have indicated that
left ventricular hypertrophy (LVH) leads to long-term harm-
ful effects and short-term benefits, while the mechanism that
regulates the transition from adaptive to maladaptive hyper-
trophy has not yet been determined [13].
* Mohammad Khaksari
mkhaksari@kmu.ac.ir
1 Physiology Research Center, Institute
ofNeuropharmacology, Kerman University ofMedical
Sciences, Kerman, Iran
2 Endocrinology andMetabolism Research Center, Kerman
University ofMedical Sciences, Kerman, Iran
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Heart Failure Reviews
1 3
Depending on heart geometry, CH can be divided into
other types, including eccentric and concentric hypertrophy.
Eccentric hypertrophy is caused by volume overload and
enhanced ventricular volume with coordinated growth in-
wall, and septal thickness indicates non-pathological eccen-
tric hypertrophy. Cardiac diseases, such as dilated cardio-
myopathy and myocardial infarction may cause pathological
eccentric hypertrophy, leading to ventricular dilatation with
cardiomyocytes preferential lengthening. Pathological con-
centric hypertrophy is characterized by an increase in the
septal thickness and free wall related to a decrease in the
left ventricular dimension. The increase in cardiomyocytes
thickness is usually greater than the length, occurring under
pathological situations including valvular heart diseases or
hypertension [14, 15]. However, non-pathological concentric
hypertrophy may be caused by exercise like wrestling [8].
Cardiac dysfunction is also dependent on other patho-
physiological conversions, such as inflammation, metabolic
derangement, apoptosis, and oxidative stress [1618]. Due
to their complexity, simple remedial methods are not suf-
ficient to manage this situation. Given that signaling path-
ways in pathological hypertrophy are created as a result of
excessive biomechanical stress or hormonal factors, there are
several overlaps between pathological CH and physiological
mechanisms.
Estrogen andits receptors (ERs)
17β-estradiol (E2) is the main female steroid hormone and
this is the main form of circulating estrogen. The other two
natural forms occurring less frequently are estrone (E1) and
estriol (E3), and the third one produced only during the preg-
nancy is estretrol (E4) [19]. In premenopausal women, the
ovaries mostly synthesize and secrete the E2. Several tissues
produce some E2, such as the brain, adipose, aortic smooth
muscle cells, vascular endothelium, and bone tissues [20].
Extragonadal generation of E2 acts locally as an intracrine
or paracrine factor in the tissue where it is synthesized, while
gonadal E2 acts mostly as an endocrine factor that affects
distal tissues [20]. Given that extragonadal E2 is still the
only source of endogenous E2 generation in postmenopausal
men and women, its generation plays a major role [20].
E2 mainly binds to classical estrogen receptors (ERs),
estrogen receptor α (ERα), and estrogen receptor β (ERβ)
[21] and non-classical estrogen receptor GPER (G protein-
coupled estrogen receptor) [22]. In general, the genomic
effect can be created by binding to ERα and ERβ [23] and
rapid non-genomic function induced by binding to GPER
[22]. It has recently been reported that in addition to intracel-
lular localization of ERα and ERβ, they are also membrane
bound. E2 binds to membrane-bound ERα, ERβ, and GPER
for rapid non-genomic function [2426].
Silent information regulator 1 (SIRT1)
Sirtuins are a preserved family of histone deacetylase
(HDACs) whose activity can extend the lifespan of many
organisms [27]. It was established, in 1999, that the human
SIRT1 gene is placed at 10q21.3 [28], encoding a protein
of amino acids747 [29]. The catalytic core contains a great
Rossmann fold domain which is described as a small zinc
finger domain and an NAD+/NADH binding protein [30].
There are NAD binding sites and near a NAD glycosylation
site, an acetylated substrate is bound to the end of the gap.
SIRT1 structure characterizes its role in cellular activi-
ties, including DNA repair, metabolic regulation, DNA rep-
lication, and gene transcription. SIRT1 uses deacetylation of
histones and a multitude of non-histone proteins to regulate a
set of cellular activities important for metabolism, apoptosis,
cell senescence, transcriptional regulation, cell growth, and
cell survival [31]. SIRT1 is the main regulator of metabo-
lism in various tissues [32]. Moreover, other targets of non-
histone SIRT1 protein include nuclear factor kappa B (NF-
κB), forkhead transcription factor O (FOXO), tumor protein
53 (p53), and peroxisome proliferator-activated receptor-γ
coactivator-1α (PGC-1α) [33, 34].
Cardioprotective mechanisms ofestrogen
andERs
It has been revealed that inflammation, apoptosis, and oxida-
tive stress are mainly involved in the mechanism responsible
for injury of many organs, especially the cardiovascular sys-
tem (Fig.1) [35]. Estrogen has protective effects in various
organs via inhibition of inflammation, apoptosis, and oxida-
tive stress [36, 37]. On the other hand, abnormal ER signal-
ing leads to the development of a number of diseases such as
cardiovascular diseases, cancer, and obesity, mainly through
inflammation, apoptosis, and oxidative stress mechanisms
[3639] (Table1).
Anti‑inflammatory actions ofestrogen
andERs
Studies suggested that estrogens have a protective effect
against heart disorders by influencing the inflammatory
response [4042] (Table1). E2 increase Cystathionine-
F-lyase (CSE) expression and reduce inflammation in the
myocardium of OVX rats [36]. It has been revealed that E2
inhibits expression of the inflammatory cytokines IL-1β
(interleukin-1β) and TNF-α (tumor necrosis factor-α) in
myocytes of old OVX [43] and in the ischemic hearts of
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Heart Failure Reviews
1 3
OVX rats [44]. Findings suggested that the activation of
ERα reduced endotoxin-induced inflammatory response
in cardiomyocyte cells [45]. In female murine hearts, ERα
confers cardioprotection after ischemia/reperfusion (I/R)
and regulates cytokine expression [46]. Estrogen reduces
TNF-α responses via ERβ activation in rat aortic smooth
muscle cells [47]. ERβ protects myocardial function fol-
lowing I/R via a reduction in IL-1β, IL-6 (interleukin-6),
and TNF-α in normal female mice hearts [48]. In addition,
G-1(a selective agonist of GPER) induced pro-survival
and anti-inflammatory effects after I/R by decreasing the
production of IL-1β, IL-6, and TNF-α [49].
Anti‑apoptotic actions ofestrogen andERs
Many growth factors, cytokines, and hormones control
apoptosis in different ways [50]. It is known that E2 reg-
ulates the balance between proliferation, cell survival,
and apoptosis via different mechanisms [51] (Table1).
E2 inhibits apoptosis-regulated signaling kinase-1 and
prevents congestive heart failure in mice [52]. Also, E2
through activation of PI3K/Akt (Phosphoinositide-3
kinase/protein kinase B) signaling leads to a reduction in
cardiomyocyte apoptosis invivo and invitro models [53].
Fig. 1 Mediatory mechanisms of estrogen and ERs in reducing
inflammation, apoptosis, and oxidative stress against cardiac dys-
function. increase; decrease; inhibit; ERα estrogen receptor α;
ERβ estrogen receptor β; GPER G protein-coupled estrogen receptor;
TNF-α tumor necrosis factor-α; IL-1β interleukin-1β; IL-6 interleu-
kin-6; CSE cystathionine-F-lyase; H2S hydrogen sulfide; CINC-2β
cytokine-induced neutrophil chemoattractant; MCP-1 monocyte che-
moattractant protein; ACAA2 acetyl coenzyme A acyltransferase 2;
ASK1 apoptosis signal-regulating kinase 1; JNK c-Jun N-terminal
kinase; p38 MAPK p38 mitogen-activated protein kinase; PI3K/Akt
phosphatidylinositol 3-kinase/protein kinase B; ROS reactive oxygen
species; GSH/GSSG glutathione/oxidized glutathione ratio; T-AOC
total antioxidant capacity; CAT catalase; SOD superoxide dismutase;
Nox4 NADPH oxidase 4; Pgts2 prostaglandin-endoperoxide synthase
2; Gpx1 glutathione peroxidase 1
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Heart Failure Reviews
1 3
Table 1 Mechanisms related to estrogen and ERs-mediated cardioprotection
Study model Effects on heart and
mechanism involved Sex Species Year Reference
Inflammation I/R animal model E2 improves cardiac
recovery due to
reduced TNF-α level
Female Rat 2006 Xu etal. [44]
Menopausal aging model E2 leads to cardioprotec-
tive HSP response due
to inhibited expression
of IL-1β and TNF-α
Female Rat 2011 Stice etal. [43]
OVX animal model E2 has cardioprotective
effects due to increased
CSE expression and
H2S generation and
decreased IL-6 and
TNF-α levels
Female Rat 2013 Zhu etal. [36]
TNF-α-induced inflam-
matory
E2 via ERβ has cardio-
protective effects due
to reduced CINC-2β
and MCP-1 mRNA
expression
Male Rat 2007 Xing etal. [47]
I/R animal model G-1 protects cardiac
function due to dimin-
ished TNF-α, IL-1β,
and IL-6
Male Rat 2010 Weil etal. [49]
I/R animal model ERα and ERβ mediates
acute myocardial pro-
tection due to reduced
TNF-α, IL-1β, and
IL-6 in females
Female and Male Mice 2006, 2008 Wang etal. [46, 48]
In vitro cell culture NG-R1 via ERα protects
cardiomyocytes due
to decreased TNF-
α, IL-1β, and IL-6
mRNA expression
H9c2 cell 2015 Zhong etal. [45]
Apoptosis Ischemia-induced
apoptosis
E2 protects the heart
against ischemic dam-
age due to reduced
caspase-3 activity and
prevented release of
cytochrome c
Female Rat 2006 Morkuniene etal. [55]
MI animal model E2 inhibits cardiomyo-
cytes apoptosis due to
activation of PI3K/Akt
signaling
Female Mice 2004 Patten etal. [53]
I/R injury in OVX
animal
ERβ improves car-
diac recovery due to
decreased cytochrome
c releaseand increased
levels of Bcl2 and
ACAA2
Female Mice 2016 Schubert etal. [57]
Gαq transgenic animal
model
E2 prevents conges-
tive heart failure due
to decreased ASK1,
JNK, and p38 MAPK
activity
Male Mice 2007 Satoh etal. [52]
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Heart Failure Reviews
1 3
E2 treatment can inhibit mitochondriadependent and ova-
riectomyinduced cardiac Fasdependent apoptotic path-
ways in OVX or menopause rat models [54]. Moreover,
E2 inhibits ischemia-induced apoptosis through reduc-
tion of caspase-3-like activity and prevents the release
of cytochrome c in female rat hearts [55]. The activation
of ERα decreased endotoxin-induced apoptosis [45] and
autophagy in cardiomyocyte cells [56]. Also, it has been
shown ERβ attenuates apoptosis and development of fibro-
sis, in pressure overload [37], and under I/R injury in OVX
mice [57]. In addition, GPER diminished cardiocyte apop-
tosis following I/R injury in myocardial cells [58].
Anti‑oxidative stress actions ofestrogen
andERs
There is a general agreement that oxidative stress leads to
cardiovascular dysfunction [59]. Estrogens have protective
effects against oxidative stress on various tissues including
the heart. E2 diminishes angiotensin II (Ang II)-induced free
radical production and stimulates the expression of super-
oxide dismutase (SOD) in vascular smooth muscle cells
[60], and in the myocardium of OVX rats [36] (Table1).
E2 reduced ROS generation in cardiomyocytes by attenuat-
ing the p38α mitogen-activated protein kinase (MAPK) and
I/R ischemia/reperfusion, HSP heat shock protein, CSE cystathionine-F-lyase, H2S hydrogen sulfide, CINC-2β cytokine-induced neutrophil che-
moattractant, MCP-1 monocyte chemoattractant protein, G-1 GPR30 agonist, NG-R1 notoginsenoside R1, ACAA2 acetylcoenzyme A acyltrans-
ferase 2, ASK1 apoptosis signal-regulating kinase 1, JNK c-Jun N-terminal kinase, p38 MAPK p38 mitogen-activated protein kinase, GSH/GSSG
glutathione/oxidized glutathione ratio, T-AOC total antioxidant capacity, CAT catalase, SOD superoxide dismutase, VSMC vascular smooth mus-
cle cells, MI myocardial infarction, ERβ−/−/TAC ERβ knockout/transverse aortic constriction, Nox4 NADPH oxidase 4, Pgts2 prostaglandin-
endoperoxide synthase 2, Gpx1 glutathione peroxidase 1
Table 1 (continued)
Study model Effects on heart and
mechanism involved Sex Species Year Reference
ERβ−/−/TAC animal
model
ERβ inhibits cardiac
hypertrophy and heart
failure due to reduced
apoptosis
Female and Male Mice 2010 Fliegner etal. [37]
In vitro cell culture NG-R1 via ERα protects
cardiomyocytes due to
decreased caspase-3
activity
H9c2 cell 2015 Zhong etal. [45]
In vitro cell culture ERα protects car-
diomyocytes due to
diminished caspase-3
activity
H9c2 cell 2018 Chen etal. [56]
In vitro cell culture G-1 prevents myocar-
dial ischemia due to
elevated Bcl-2 levels,
and decreased Bax
H9c2 cell 2015 Li etal. [58]
Oxidative stress OVX animal model E2 has cardioprotective
effects due to increased
GSH/GSSG ratio,
T-AOC, CAT, and
SOD activity
Female Rat 2013 Zhu etal. [36]
Ethanol-induced myo-
cardial oxidative stress
ERα inhibits myocardial
dysfunction due to
decreased ROS genera-
tion
Female Rat 2016 Yao and Abdel-Rahman
[62]
GPER knockout model E2 has cardioprotective
effects due to reduced
GSH/GSSG ratio and
Nox4, Pgts2, and Gpx1
genes expression
Female Mice 2018 Wang etal. [63]
In vitro cell culture E2 has vasoprotective
effects due to increased
MnSOD mRNA
expression
VSMCs cell 2003 Strehlow etal. [60]
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Heart Failure Reviews
1 3
enhancing the p38β MAPK activity [61]. Also, it has been
reported that E2 and ERα inhibit ethanol-induced myocar-
dial oxidative stress in female rats [62]. Findings have shown
that GPER KO leads to enhancement of cardiac oxidative
stress measured as a reduced ratio of glutathione to oxidized
glutathione (GSH/GSSG) and increased oxidative stress-
related genes expression in mice [63].
Estrogen andERs effects againstCH
Several studies have shown the effect of gender in cardiovas-
cular diseases [64, 65], as it is less probable, compared to men,
that premenopausal women will suffer cardiovascular diseases
including hypertrophy [66, 67]. Sex differences during cardiac
pathological hypertrophy can be related to sex hormones or
their receptors [68]. Hypertension or other non-ischemic factors
are the most common reason for heart failure in women, espe-
cially in postmenopausal situations [69, 70]. The mechanisms
of estrogens’ cardioprotective role have been studied, and it
seems that both classical ERs are involved to some extent [71].
Endothelial cells, mostly express ERα, while ERβ stimulates
nitric oxide production. Hence, vascular wall dilation, which
has useful hypotensive effects, activates both receptors [72, 73].
It has been indicated that plasma membrane is linked to
the non-nuclear fraction of ERα which is responsible for vas-
cular functions resulting from estrogens, including endothe-
lial NO production, endothelial repair acceleration, and rapid
dilatation [74]. In addition, Ang II activation has an incre-
ment effect on myocyte hypertrophy via calcineurin induction,
expression of β-MHC, the activation of extracellular signal-
regulated kinase (ERK), and mitogen-activated protein kinases
(MAPK). Inhibition of Ang II and its signaling pathway can
cause E2-induced protection of cardiac muscle [75]. Also,
estrogen inhibits salt-induced CH development in proANP
(atrial natriuretic peptide) gene-disrupted mice [76] (Table2).
Both classical ERs participate in cardioprotective effects of
estrogen on CH [77, 78] through the following mechanisms:
blocking of the enhanced p38-MAPK phosphorylation, increas-
ing the expression of ANP, suppressing calcineurin/NF-AT3
signaling pathways, PI3K/Akt upregulation [7981], lowered
peripheral artery resistance and reduced systolic blood pressure
[82]. Moreover, activation of GPER and G-1 administration
inhibits Aldo-induced hypertrophy and Ang II-induced hyper-
trophy in cardiac myocytes [83, 84] via the activation of the
PI3K-Akt-mTOR signaling pathway [85] (Table2).
Cardioprotective mechanisms ofSIRT1
SIRT1 expression is abundant in mammalian hearts, and
many cell functions are regulated by the deacetylation of
histones and some non-histone proteins [86]. SIRT1 as a
cardioprotective molecule is frequently found in the nucleus
and is widely considered in the cardiovascular system, tak-
ing part in intracellular signaling in the heart [87] (Table3).
Evidence represents the preventive role of SIRT1 in heart
injuries (Fig.2) [88, 89], such as dilated cardiomyopathy
in patients and post-myocardial infarction in rats [90], oxi-
dative stress, and ischemia/reperfusion (I/R) injury, hyper-
trophy, and cardiomyocyte apoptosis [9193]. In addition,
SIRT1 is recognized as an anti-atherosclerosis factor due to
its anti-inflammatory, anti-apoptotic, and antioxidant activi-
ties in the endothelium [9395].
SIRT1 anti‑inflammatory effects
There is a pro-inflammatory profile in the vasculature of
animals and humans specified by an increase in the num-
ber of chemokines and cytokines [96, 97]. Furthermore,
inflammation is the main mechanism of cardiovascular
disease [98]. Therefore, inflammation results in prevent-
ing the progression of cardiovascular diseases [99]. Various
studies have indicated that SIRT1 can decrease inflamma-
tion caused by heart failure, and myocardial hypertrophy
[100, 101]. Moreover, it has been confirmed that dioscin
protects against coronary heart disease (CHD) by reducing
inflammation via SIRT1/Nrf2 and p38 MAPK pathways
[102]. The PE-induced increment in CH is also prevented
by SIRT1 in mRNA levels of the pro-inflammatory cytokine
monocyte chemoattractant protein-1 (MCP-1) and also it
inhibits the increased nuclear factor-kB (NF-kB) activity
associated with PE exposure [100]. In addition, the observa-
tions have revealed SIRT1’s inhibitory role against CH in
the SIRT1–PPARα interaction [100] because invitro evi-
dence has shown that RSV in PPARα-null mice does not
inhibit isoproterenol-induced CH and inflammation [100].
It has been reported that Kallistatin (KS, a serine protease
inhibitor) by promoting the SIRT1 expression decreases
the inflammation of myocardial tissue in heart failure rats
[103].
SIRT1 anti‑apoptotic effects
Apoptosis involves complex mechanisms. Interactions
between apoptotic factors and SIRT1 may be associated
with subcellular localization. Transcription factors like
FOXO, Ku70, and p53 are firstly deacetylate by SIRT1 in
the nucleus, to suppress responses to downstream apoptosis
cell signaling and thus prevent cell death [104].
It has been revealed that heart failure, age-dependent CH,
and apoptosis can be reduced by increasing the concentra-
tion of SIRT1 in the heart [87]. SIRT1 also suppresses the
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Heart Failure Reviews
1 3
activity of pro-apoptotic p53 and regulates the activity of
p53 via deacetylation [105], thereby decreasing cardiomyo-
cyte apoptosis [106]. Inhibition of SIRT1 causes an increase
in acetylation level and p53 activity, so leading to cardio-
myocytes’ death and heart failure [107, 108]. The results
represent that when cellular apoptosis is caused by oxida-
tive stress, SIRT1 expression increases to prevent apoptosis
and preserve myocardiocytes [109]. SIRT1 inhibited cellu-
lar injury in myocardial infarction by activating Bcl-xL and
thioredoxin-1, and repressing activities of Bax and cleaved
caspase-3 [92]. In addition, it has been demonstrated that
KS via promoting the SIRT1 expression diminished Bax
and caspase-3/9 activities [103]. It has been revealed that
RSV protects cardiomyocytes against hypoxia-induced
apoptosis via miR-30d-5p/SIRT1/NF-κB axis [110] and
SIRT1–FOXO1 pathway [93].
SIRT1 anti‑oxidative stress effects
The overproduction of reactive oxygen species (ROS)
including oxygen free radicals and related entities, such
as peroxynitrite, singlet oxygen, nitric oxide (NO), hydro-
gen peroxide (H2O2), and superoxide free radicals, leads
to oxidative stress [111]. Various studies have shown the
destructive effects of oxidative stress on vascular and car-
diac aging [112, 113]. Other findings have revealed that
oxidative stress is involved in CH and other cardiovascular
diseases [114]. SIRT1 is considered a strong intracellular
inhibitor of oxidative stress and inflammatory responses.
The probable role of SIRT1 against cardiovascular disease
has been considered due to its inhibitory effect against
oxidative stress [114]. SIRT1 and IGF-1 are the two main
regulators of cardiomyocyte cardiac stress [96, 112]. IGF-1
uses SIRT1 to protect the heart against oxidative stress
[113]. Endothelial cells pretreatment with RSV increases
SIRT1 levels and activity, leading to less nitric oxide syn-
thase acetylation [98] (Table3). Another study has shown
the protective effects of RSV on endothelial blood vessels
[115].
Overexpression of SIRT1 protected against oxidative stress
via CH, cardiac dysfunction, apoptosis/fibrosis, and induced
the expression of catalase [87]. Crosstalk between the ROS
signaling and SIRT1 stimulates the autophagy reduction
response in the mice cardiac muscle [87, 116]. It has been
established that the increment in the catalase expression is
induced via the FOXO1 transcription factor. SIRT1 inactivates
FOXO factors through the deacetylation of these factors and
also stimulates antioxidants expression (MnSOD and catalase)
and enhances SIRT1 expression via an auto-feedback loop [87,
Table 2 Protective effects of estrogen and ERs against CH
ANP atrial natriuretic peptide, BNP brain natriuretic peptide, TAC transverse aortic constriction, p38-MAPK p38 mitogen-activated protein
kinase, βMHC beta-myosin heavy chain, SHR spontaneously hypertensive rats, AAC abdominal aortic constriction, G-1 GPR30 or GPER ago-
nist, AngII angiotensin II, Ald aldosterone, NRCM neonatal cardiac ventricular myocytes, CH cardiac hypertrophy
Study model Effect on the heart and mechanism involved Sex Species Year References
SHR animal model ERβ agonist 8β-VE2 diminishes hypertrophy
due to reduced cardiac afterload
Female Rat 2008 Jazbutyte etal. [82]
AAC-induced CH E2 decreases CH due to suppressed cal-
cineurin/NF-AT3 signaling pathways and
up-regulating PI3K/Akt
Female Rat 2005 Wu etal. [80]
SHR animal model E2 or 16α-LE2 agonist attenuates CH due to
increased cardiac output, α-MHC, and ANP
expression
Female Rat 2005 Pelzer etal. [78]
SHR animal model G-1 inhibits CH due to diminished ANP and
BNP mRNA expression
Male Rat 2020 Di Mattia etal. [83]
ANP + / − animal model E2 inhibits hypertrophy due to reduced ANP
and BNP mRNA expression
Male and female Mice 2007 Sangaralingham etal. [76]
ER − / − /TAC induced model E2 via ERβ prevents CH due to prohibited
p38-MAPK phosphorylation and elevated
ANP expression
Female Mice 2006 Babiker etal. [79]
TAC-induced CH E2 and 16α-LE2 agonist suppress myocardial
hypertrophy due to decreased BNP, ANP,
and β-MHC
Female Mice 2012 Westphal etal. [81]
AngII-induced hypertrophy E2 through ERβ reduces CH due to stimu-
lated BNP and decreased α/β MHC ratio
andinhibited calcineurin activity
Female Mice 2008 Pedram etal. [75]
Ald-induced hypertrophy G-1 inhibits CH due to decreased BNP pro-
tein expression and BNP protein/DNA ratio
NRCM cell 2020 Di Mattia etal. [83]
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Heart Failure Reviews
1 3
117]. So, it reduces the deterioration of cardiac function and
hypertrophy, suppresses fibrosis, and improves survival [90,
117, 118].
SIRT1 controls the ROS production through NF-κB sign-
aling [119, 120] and in this way reduces the expression of
inducible nitric oxide synthase (iNOS) and increases genera-
tion of reactive nitrogen radicals [121]. Moreover, by increas-
ing endothelial nitric oxide synthase (eNOS), SIRT1 can
enhance cardiac tolerance to oxidative stress and ischemia
[122] (Table3).
SIRT1 effects againstCH
The studies indicate that SIRT1 plays an extensive role
against CH and other cardiovascular deficits. Control of
endothelial angiogenic activities during vascular growth
[123] and abnormal cardiac growth in the SIRT1-knockout
mouse model [124] demonstrate the key role of SIRT1
in heart formation [125]. Also, under stress condi-
tions (e.g., starvation), SIRT1 increased cardiomyocyte
growth because inhibition of SIRT1 function leading to
Table 3 Mechanisms related to SIRT1-mediated cardioprotection
CHD coronary heart disease, DOX doxorubicin, HF heart failure, KS kallistatin, PE phenylephrine, NCM neonatal cardiomyocyte, ISO isopro-
terenol, MCP-1 monocyte chemoattractant protein-1, Tg-SIRT1 SIRT1 transgenic mice, mIGF-1 insulin-like growth factor-1 isoform, TBARS
thiobarbituric acid reactive substances, HUVECs human umbilical vein endothelial cells, eNOS endothelial nitric oxide synthase, Mn-SOD man-
ganese superoxide dismutase, ROS reactive oxygen spices, RSV resveratrol, PO pressure overload
Study model Effect on the heart and mecha-
nism involved Sex Species Year References
Inflammation CHD model induction Dioscin via SIRT1 protects against
CHD due to reducedTNF-α,
IL-1β, IL-6, and IL-18 levels
Male Pig 2018 Yang etal. [102]
DOX-induced HF KS via SIRT1 inhibits HF due to
reducedTNF-α, IL-1β, IL-6, and
IL-18 levels
Male Rat 2020 Xie etal. [103]
ISO-induced hypertrophy SIRT1–PPARα interaction inhibits
CH due to decreased NF-kB and
MCP-1 mRNA levels
Male Rat 2011 Planavila etal. [100]
In vitro cell culture SIRT1–PPARα interaction inhibits
CH due to decreased NF-kB and
MCP-1 mRNA levels
NCM cell 2011 Planavila etal. [100]
Apoptosis I/R animal model SIRT1 protects the heart against
I/R due to activated Bcl-xL and
thioredoxin-1 and suppressed
cleaved caspase 3 activity and Bax
through the upregulation of FOXO
Male Mice 2010 Hsu etal. [92]
Cardiac-specific Tg-SIRT1 SIRT1 prevents cardiac dysfunction
due to elevated Bcl-2 and Bcl-xL
expression
Male Mice 2007 Alcendor etal. [87]
DOX-induced HF KS via SIRT1 inhibits heart failure
due to reduced Bax and cas-
pase3/9 activity
Male Rat 2020 Xie etal. [103]
In vitro cell culture RSV protects cardiomyocytes due
to reduced cleaved caspase 3
H9c2 cell 2020 Han etal. [110]
Oxidative stress Cardiac-specific Tg- SIRT1 SIRT1 prevents heart failure due to
increased mIGF-1 expression
Male Mice 2012 Bolasco etal. [113]
PO-induced CH RSV via SIRT1 prevents CH due to
reduced TBARS
Male Rat 2010 Wojciechowski etal. [129]
SIRT1-induced Mn-SOD RSV via SIRT1 prevents heart fail-
ure due to increased Mn-SOD
Male Hamster 2010 Tanno etal. [90]
In vitro cell culture SIRT1 inhibits endothelial dysfunc-
tion due to eNOS acetylation
HUVECs 2010 Arunachalam etal. [98]
In vitro cell culture SIRT1 promotes vascular tone due
to eNOS deacetylation
Endothelial cell 2007 Mattagajasingh etal. [122]
In vitro cell culture RSV via SIRT1 protects the heart
due to decreased ROS and
increased Mn-SOD
C2C12 cells 2010 Tanno etal. [90]
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Heart Failure Reviews
1 3
cardiomyocyte death [107] (Table4). It has been demon-
strated that SIRT1 leads to resistance to CH increase in
response to both physiologic and pathologic hypertrophic
stimuli [126]. The study showed that a low to moderate
SIRT1 expression has protective effects against cardiac
dysfunction, apoptosis, and age-related CH [87]. Addi-
tionally, SIRT1 overexpression inhibits cardiac dysfunc-
tion and age-dependent CH [87, 127]. Findings show that
mIGF1 (insulin-like growth factor-1 isoform) protects
cardiomyocytes against CH via SIRT1 activity [91]. The
results regarding the beneficial effects of SIRT1 on CH are
caused by the following mechanism(s):
Inhibition of poly (ADP-ribose) polymerases 2 (PARP-2)
protein and mRNA levels [128] reduce the oxidative stress
by increasing the antioxidants expression (e.g., catalase)
[87, 129] (Tables3 and 4), increase the ability of fibro-
blast growth factor 21 (FGF21) [130], activate adenosine
monophosphate-activated protein kinase (AMPK) [131]
(Table4), decrease the mRNA expressions of ANP and
β-MHC (β-myosin heavy chain) [131], and activate Akt
[132, 133].
SIRT1 andestrogen crosstalk
Studies revealed that SIRT1 is necessary for modulating
ERα-signaling pathways (Fig.3). HDACs are directly asso-
ciated with ERα protein, regulating its downstream gene
transcription [134, 135]. ERα and its downstream gene tran-
scription are suppressed by inhibiting SIRT1 activity [136].
On the other hand, SIRT1 is a unified player activated by
estrogens, via ERs [137].
It has been shown that cardiovascular dysfunction in post-
menopausal metabolic syndrome (PMS) is inhibited by E2
through its effect on SIRT1/AMPK/H3 acetylation. Downregu-
lation of SIRT1/AMPK/H3 acetylation in the aorta and heart is
related to cardiac apoptosis induced by PMS. Ang II-induced
contractions and H3 acetylation are partially suppressed by
Fig. 2 Mediatory mechanisms of SIRT1 in reducing inflammation,
apoptosis, and oxidative stress against cardiac dysfunction. increase;
decrease; inhibit; SIRT1 silent information regulator 1; TNF-α
tumor necrosis factor-α; IL-1β interleukin-1β; IL-6 interleukin-6; IL-
18 interleukin-18; MCP-1 monocyte chemoattractant protein; NF-kB
nuclear factor kappa B; Bcl-xL B cell lymphoma-extra-large; TBARS
thiobarbituric acid reactive substances; eNOS endothelial nitric oxide
synthase, ROS reactive oxygen species; mIGF‐1 insulin-like growth
factor-1 isoform; MnSOD manganese superoxide dismutase
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Heart Failure Reviews
1 3
Table 4 Protective effects of SIRT1 against CH
AAC abdominal aortic constriction, ANF atrial natriuretic factor, BNP brain natriuretic peptide, β-MHC β-myosin heavy chain, PARP-2 poly
(ADP-ribose) polymerase-2, PO pressure overload, TBARS thiobarbituric acid reactive substances, ANP atrial natriuretic peptide, AMPK adeno-
sine monophosphate-activated protein kinase, Tg-SIRT1 SIRT1 transgenic, CH cardiac hypertrophy, Ang II angiotensin II, FGF21 fibroblast
growth factor 21, PE phenylephrine, TAC transverse aortic constriction, Bcl-xL B cell lymphoma-extra-large, mIGF‐1 Insulin-like growth fac-
tor-1 isoform, ACTA-1 skeletal muscle alpha-actin gene, MDA malondialdehyde, ROS reactive oxygen species, UCP-1 uncoupling protein-1, MT-
2 melatonin receptor 2, MCP-1 monocyte chemoattractant protein-1, NF-kB nuclear factor kappa B
Study model Effect on the heart and mechanism involved Sex Species Year References
AAC-induced hypertrophy SIRT1 prevents CH due to decreased ANF, BNP, β-MHC
mRNA and PARP-2 mRNA and protein expression
Male Rat 2013 Geng etal. [128]
PO-induced CH RSV via SIRT1 prevents CH due to reduced TBARS Male Rat 2010 Wojciechowski etal. [129]
TAC- induced CH SIRT1 inhibits CH due to decreased ANP and β-MHC
mRNA expressions by AMPK activation
Male Rat 2018 Dong etal. [131]
Ang II-induced CH SIRT1 inhibits CH due to increased FGF21 Male Mice 2019 Li etal. [130]
Cardiac-specific Tg-SIRT1 SIRT1 prevents CH due to elevated Bcl-2 and Bcl-xL
expression
Male Mice 2007 Alcendor etal. [87]
In vitro cell culture SIRT1 inhibits CH due to decreased ANP and β-MHC
mRNA expressions by AMPK activation
Myocardial cell 2018 Dong etal. [131]
In vitro cell culture SIRT1–PPARα interaction inhibits CH due to decreased
NF-kB and MCP-1 mRNA levels
NCM cell 2011 Planavila etal. [100]
In vitro cell culture SIR2α inhibits CH due to decreased ANF mRNA expres-
sion, cleaved caspase 3 activity and p53 acetylation
NRVMs 2004 Alcendor etal. [107]
In vitro cell culture SIRT1 via mIGF1 inhibits CH due to diminished ANP,
BNP, ACTA-1 mRNA, and ROS, MDA protein levels
and increased adiponectin, UCP-1, and MT-2 mRNA
expression
HL1 cell 2010 Vinciguerra etal. [91]
Fig. 3 Probable interaction between estrogen and SIRT1 against car-
diac hypertrophy. Inflammation, apoptosis, and oxidative stress seem
to be among the most important mechanisms involved in the devel-
opment of myocardial hypertrophy. On the other hand, estrogen can
reduce or inhibit oxidative stress, apoptosis, and inflammation by
regulating the expression of SIRT1 protein, thereby reducing the inci-
dence of pathological CH. increase; inhibit; ERα estrogen recep-
tor α; SIRT1 silent information regulator 1, GPER G protein-coupled
estrogen receptor
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Heart Failure Reviews
1 3
E2, and they also restore the SIRT1/P-AMPK protein expres-
sion [138]. Estrogen therapy elevates the P-AMPK and SIRT1
expression, supporting the opinion that E2 could have cardio-
protective effects on PMS [139, 140]. It has been reported that
E2 inhibits brain ischemic stroke and cardiac ischemia through
SIRT1/P-AMPK signaling [141, 142].
It has been demonstrated that E2 can increase SIRT1
expression and reduce PPARγ and adipogenesis in aged
mice and in cell culture models [143, 144]. Moreover, SIRT1
is upstream of ERα and it is responsible for promoting the
expression of ERα [136]. On the other hand, ERα is upstream
of SIRT1 in breast cancer cells and binds to the SIRT1 pro-
moter to induce SIRT1 gene expression [145]. SIRT1 expres-
sion in white adipose tissue (WAT) from ERα knockout mice
was reduced, indicating that ERα may act as an upstream of
SIRT1 [146]. In line with that, SIRT1 is upstream of ERα, it
has been shown ERα protein is decreased by SIRT1 knockout
and estrogen-responsive genes are eventually reduced [136].
Findings have been revealed that E2 and the selective GPER
agonist G-1 induce SIRT1 expression in cancer cells through
the rapid activation of the EGFR signaling [147].
Conclusions
CH is an important critical component of cardiac remod-
eling. Knowledge of CH has advanced dramatically over the
past decade and various modifiers involved in hypertrophy
and important signaling molecules have been identified. The
main regulator of physiological processes is estrogen, and
its deficiency is associated with CH and other cardiovascu-
lar diseases and leads to a disease-promoting state in post-
menopausal women. Given that the increase in SIRT1 has
a protective role in cardiomyocyte injury during CH, since
there is a crosstalk between estrogen and SIRT1, it can be
said that estrogen may improve the prognosis of CH and
protect the heart from injury by modulating SIRT1 signaling
and inhibiting oxidative stress, inflammation, and apoptosis.
Therefore, it is recommended that SIRT1 be used as a new
treatment strategy alone or in combination with estrogen in
postmenopausal women with cardiac hypertrophy.
Authors’ contributions ZH and MK reviewed the literature and wrote
the paper. Both authors read and approved the final manuscript.
Funding This research did not receive any specific grant from funding
agencies in the public, commercial, or not-for-profit sectors.
Declarations
Ethics approval and consent to participate This article does not contain
any studies with human participants or animals performed by any of
the authors.
Conflicts of interest The authors declare that there are no conflicts of
interest.
References
1. Patrizio M, Marano G (2016) Gender differences in cardiac
hypertrophic remodeling. Ann Ist Super Sanita 52(2):223–229
2. Di Minno A, Stornaiuolo M, NovellinoE (2019) Molecular
Scavengers, Oxidative Stress and Cardiovascular Disease. J Clin
Med8(11)
3. Zhou L, Ma B, Han X (2016) The role of autophagy in angio-
tensin II-induced pathological cardiac hypertrophy. J Mol Endo-
crinol 57(4):R143-r152
4. McMullen JR, Jennings GL (2007) Differences between patho-
logical and physiological cardiac hypertrophy: novel therapeu-
tic strategies to treat heart failure. Clin Exp Pharmacol Physiol
34(4):255–262
5. Berenji K etal (2005) Does load-induced ventricular hypertro-
phy progress to systolic heart failure? Am J Physiol Heart Circ
Physiol 289(1):H8-h16
6. Frank D etal (2008) Gene expression pattern in biomechanically
stretched cardiomyocytes: evidence for a stretch-specific gene
program. Hypertension 51(2):309–318
7. Weeks KL, McMullen JR (2011)The athlete's heart vs. the fail-
ing heart: can signaling explain the two distinct outcomes? Physi-
ology (Bethesda)26(2):97–105
8. Bernardo BC etal (2010) Molecular distinction between physio-
logical and pathological cardiac hypertrophy: experimental find-
ings and therapeutic strategies. Pharmacol Ther 128(1):191–227
9. Perrino C etal (2006) Intermittent pressure overload triggers
hypertrophy-independent cardiac dysfunction and vascular rar-
efaction. J Clin Invest 116(6):1547–1560
10. Lyon RC etal (2015) Mechanotransduction in cardiac hypertro-
phy and failure. Circ Res 116(8):1462–1476
11. Francis GS, McDonald KM, CohnJN (1993) Neurohumoral acti-
vation in preclinical heart failure. Remodeling and the potential
for intervention. Circulation87(5 Suppl):Iv90–6
12. Maillet M, van Berlo JH, Molkentin JD (2013) Molecular basis
of physiological heart growth: fundamental concepts and new
players. Nat Rev Mol Cell Biol 14(1):38–48
13. Schiattarella GG, Hill JA (2015) Inhibition of hypertrophy is a
good therapeutic strategy in ventricular pressure overload. Cir-
culation 131(16):1435–1447
14. Selby DE etal (2011) Tachycardia-induced diastolic dysfunction
and resting tone in myocardium from patients with a normal ejec-
tion fraction. J Am Coll Cardiol 58(2):147–154
15. Heineke J, Molkentin JD (2006) Regulation of cardiac hypertro-
phy by intracellular signalling pathways. Nat Rev Mol Cell Biol
7(8):589–600
16. Shimizu I, Minamino T (2016) Physiological and pathological
cardiac hypertrophy. J Mol Cell Cardiol 97:245–262
17. Zhang WX etal (2019) Melatonin protects against sepsis-induced
cardiac dysfunction by regulating apoptosis and autophagy via
activation of SIRT1 in mice. Life Sci 217:8–15
18. García N, Zazueta C, Aguilera-Aguirre L (2017) Oxidative Stress
and Inflammation in Cardiovascular Disease. Oxid Med Cell
Longev 2017:5853238
19. Coelingh Bennink HJT etal (2017) Pharmacodynamic effects of
the fetal estrogen estetrol in postmenopausal women: results from
a multiple-rising-dose study. Menopause24(6):677–685
20. Simpson ER (2003) Sources of estrogen and their importance. J
Steroid Biochem Mol Biol 86(3–5):225–230
21. Sirianni R etal (2008) The novel estrogen receptor, G protein-
coupled receptor 30, mediates the proliferative effects induced
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Heart Failure Reviews
1 3
by 17beta-estradiol on mouse spermatogonial GC-1 cell line.
Endocrinology 149(10):5043–5051
22. Amirkhosravi L etal (2021) E2-BSA and G1 exert neuroprotec-
tive effects and improve behavioral abnormalities following trau-
matic brain injury: The role of classic and non-classic estrogen
receptors. Brain Res1750:147168
23. Deschamps AM, Murphy E, Sun J (2010) Estrogen receptor
activation and cardioprotection in ischemia reperfusion injury.
Trends Cardiovasc Med 20(3):73–78
24. Bopassa JC etal (2010) A novel estrogen receptor GPER
inhibits mitochondria permeability transition pore opening and
protects the heart against ischemia-reperfusion injury. Am J
Physiol Heart Circ Physiol 298(1):H16-23
25. Babiker FA etal (2004) 17beta-estradiol antagonizes cardio-
myocyte hypertrophy by autocrine/paracrine stimulation of a
guanylyl cyclase A receptor-cyclic guanosine monophosphate-
dependent protein kinase pathway. Circulation 109(2):269–276
26. Grohé C etal (1997) Cardiac myocytes and fibroblasts contain
functional estrogen receptors. FEBS Lett 416(1):107–112
27. Darvishzadeh Mahani F, Khaksari M, Raji-Amirhasani A
(2021)Renoprotective effects of estrogen on acute kidney
injury: the role of SIRT1. Int Urol Nephrol
28. Alcaín FJ, Villalba JM (2009) Sirtuin inhibitors. Expert Opin
Ther Pat 19(3):283–294
29. Voelter-Mahlknecht S, Mahlknecht U (2006) Cloning, chro-
mosomal characterization and mapping of the NAD-dependent
histone deacetylases gene sirtuin 1. Int J Mol Med 17(1):59–67
30. Lawson M etal (2010) Inhibitors to understand molecular
mechanisms of NAD(+)-dependent deacetylases (sirtuins).
Biochim Biophys Acta 1799(10–12):726–739
31. Finkel T, Deng CX, Mostoslavsky R (2009) Recent pro-
gress in the biology and physiology of sirtuins. Nature
460(7255):587–591
32. Ma L, Li Y (2015) SIRT1: role in cardiovascular biology. Clin
Chim Acta 440:8–15
33. Brunet A etal (2004) Stress-dependent regulation of FOXO
transcription factors by the SIRT1 deacetylase. Science
303(5666):2011–2015
34. Rodgers JT etal (2005) Nutrient control of glucose homeo-
stasis through a complex of PGC-1alpha and SIRT1. Nature
434(7029):113–118
35. Darband SG etal (2020) Combination of exercise training
and L-arginine reverses aging process through suppression of
oxidative stress, inflammation, and apoptosis in the rat heart.
Pflugers Arch 472(2):169–178
36. Zhu X etal (2013) Estrogens increase cystathionine-γ-lyase
expression and decrease inflammation and oxidative stress
in the myocardium of ovariectomized rats. Menopause
20(10):1084–1091
37. Fliegner D etal (2010) Female sex and estrogen receptor-beta
attenuate cardiac remodeling and apoptosis in pressure overload.
Am J Physiol Regul Integr Comp Physiol 298(6):R1597–R1606
38. Jordan VC (2015) The new biology of estrogen-induced apop-
tosis applied to treat and prevent breast cancer. Endocr Relat
Cancer 22(1):R1-31
39. Macciò A, Madeddu C (2011) Obesity, inflammation, and post-
menopausal breast cancer: therapeutic implications. Sci World J
11:2020–2036
40. Störk S etal (2004) Estrogen, inflammation and cardiovascular
risk in women: a critical appraisal. Trends Endocrinol Metab
15(2):66–72
41. Pradhan AD etal (2002) Inflammatory biomarkers, hormone
replacement therapy, and incident coronary heart disease: pro-
spective analysis from the Women’s Health Initiative observa-
tional study. JAMA 288(8):980–987
42. Danesh J etal (2000) Low grade inflammation and coronary
heart disease: prospective study and updated meta-analyses. BMJ
321(7255):199–204
43. Stice JP etal (2011) 17β-Estradiol, aging, inflammation,
and the stress response in the female heart. Endocrinology
152(4):1589–1598
44. Xu Y etal (2006) Estrogen improves cardiac recovery after
ischemia/reperfusion by decreasing tumor necrosis factor-alpha.
Cardiovasc Res 69(4):836–844
45. Zhong L etal (2015) Estrogen receptor α mediates the effects
of notoginsenoside R1 on endotoxin-induced inflammatory and
apoptotic responses in H9c2 cardiomyocytes. Mol Med Rep
12(1):119–126
46. Wang M etal (2006) Estrogen receptor-alpha mediates acute
myocardial protection in females. Am J Physiol Heart Circ Phys-
iol 290(6):H2204–H2209
47. Xing D etal (2007) Estrogen modulates TNF-alpha-induced
inflammatory responses in rat aortic smooth muscle cells through
estrogen receptor-beta activation. Am J Physiol Heart Circ Phys-
iol 292(6):H2607–H2612
48. Wang M etal (2008) Estrogen receptor beta mediates acute myo-
cardial protection following ischemia. Surgery 144(2):233–238
49. Weil BR etal (2010) Signaling via GPR30 protects the myocar-
dium from ischemia/reperfusion injury. Surgery 148(2):436–443
50. Kiess W, Gallaher B (1998) Hormonal control of programmed
cell death/apoptosis. Eur J Endocrinol 138(5):482–491
51. Altucci L etal (1996) 17beta-Estradiol induces cyclin D1 gene
transcription, p36D1-p34cdk4 complex activation and p105Rb
phosphorylation during mitogenic stimulation of G(1)-arrested
human breast cancer cells. Oncogene 12(11):2315–2324
52. Satoh M etal (2007) Inhibition of apoptosis-regulated signaling
kinase-1 and prevention of congestive heart failure by estrogen.
Circulation 115(25):3197–3204
53. Patten RD etal (2004) 17beta-estradiol reduces cardiomyo-
cyte apoptosis invivo and invitro via activation of phospho-
inositide-3 kinase/Akt signaling. Circ Res 95(7):692–699
54. Liou CM etal (2010) Effects of 17beta-estradiol on cardiac apop-
tosis in ovariectomized rats. Cell Biochem Funct 28(6):521–528
55. Morkuniene R, Arandarcikaite O, Borutaite V (2006) Estradiol
prevents release of cytochrome c from mitochondria and inhib-
its ischemia-induced apoptosis in perfused heart. Exp Gerontol
41(7):704–708
56. Chen BC etal (2018) Estrogen and/or estrogen receptor α inhibits
BNIP3-induced apoptosis and autophagy in H9c2 cardiomyoblast
cells. Int J Mol Sci19(5)
57. Schubert C etal (2016) Reduction of apoptosis and preservation
of mitochondrial integrity under ischemia/reperfusion injury is
mediated by estrogen receptor β. Biol Sex Differ 7:53
58. Li WL, Xiang W, Ping Y (2015) Activation of novel estrogen
receptor GPER results in inhibition of cardiocyte apoptosis and
cardioprotection. Mol Med Rep 12(2):2425–2430
59. Arias-Loza PA, Muehlfelder M, Pelzer T (2013) Estrogen and
estrogen receptors in cardiovascular oxidative stress. Pflugers
Arch 465(5):739–746
60. Strehlow K etal (2003) Modulation of antioxidant enzyme
expression and function by estrogen. Circ Res 93(2):170–177
61. Kim JK etal (2006) Estrogen prevents cardiomyocyte apoptosis
through inhibition of reactive oxygen species and differential reg-
ulation of p38 kinase isoforms. J Biol Chem 281(10):6760–6767
62. Yao F, Abdel-Rahman AA (2016) Estrogen receptor ERα plays
a major role in ethanol-evoked myocardial oxidative stress and
dysfunction in conscious female rats. Alcohol 50:27–35
63. Wang H etal (2018) G protein-coupled estrogen receptor (GPER)
deficiency induces cardiac remodeling through oxidative stress.
Transl Res 199:39–51
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Heart Failure Reviews
1 3
64. Mosca L, Barrett-Connor E, Wenger NK (2011) Sex/gender dif-
ferences in cardiovascular disease prevention: what a difference
a decade makes. Circulation 124(19):2145–2154
65. Rodgers JL etal (2019) Cardiovascular risks associated with
gender and aging. J Cardiovasc Dev Dis6(2)
66. Kander MC, Cui Y, Liu Z (2017) Gender difference in oxidative
stress: a new look at the mechanisms for cardiovascular diseases.
J Cell Mol Med 21(5):1024–1032
67. Czubryt MP etal (2006) The role of sex in cardiac function and
disease. Can J Physiol Pharmacol 84(1):93–109
68. Donaldson C etal (2009) Estrogen attenuates left ventricu-
lar and cardiomyocyte hypertrophy by an estrogen receptor-
dependent pathway that increases calcineurin degradation. Circ
Res104(2):265–75, 11p following 275
69. de Kat AC etal (2017) Unraveling the associations of age and
menopause with cardiovascular risk factors in a large popula-
tion-based study. BMC Med 15(1):2
70. Regitz-Zagrosek V et al (2010) Sex and gender differ-
ences in myocardial hypertrophy and heart failure. Circ J
74(7):1265–1273
71. Murphy E (2011) Estrogen signaling and cardiovascular dis-
ease. Circ Res 109(6):687–696
72. Guo X etal (2005) Estrogen induces vascular wall dilation:
mediation through kinase signaling to nitric oxide and estrogen
receptors alpha and beta. J Biol Chem 280(20):19704–19710
73. Zhu Y etal (2002) Abnormal vascular function and hyper-
tension in mice deficient in estrogen receptor beta. Science
295(5554):505–508
74. Adlanmerini M etal (2014) Mutation of the palmitoylation site
of estrogen receptor α invivo reveals tissue-specific roles for
membrane versus nuclear actions. Proc Natl Acad Sci U S A
111(2):E283–E290
75. Pedram A etal (2008) Estrogen inhibits cardiac hypertrophy:
role of estrogen receptor-beta to inhibit calcineurin. Endocri-
nology 149(7):3361–3369
76. Sangaralingham SJ, Tse MY, Pang SC (2007) Estrogen protects
against the development of salt-induced cardiac hypertrophy
in heterozygous proANP gene-disrupted mice. J Endocrinol
194(1):143–152
77. Tsai CY etal (2017) E2/ER β inhibit ISO-induced cardiac cel-
lular hypertrophy by suppressing Ca2+-calcineurin signaling.
PLoS One12(9):e0184153
78. Pelzer T etal (2005) The estrogen receptor-alpha agonist
16alpha-LE2 inhibits cardiac hypertrophy and improves hemo-
dynamic function in estrogen-deficient spontaneously hyper-
tensive rats. Cardiovasc Res 67(4):604–612
79. Babiker FA etal (2006) Estrogen receptor beta protects the
murine heart against left ventricular hypertrophy. Arterioscler
Thromb Vasc Biol 26(7):1524–1530
80. Wu CH etal (2005) 17beta-estradiol reduces cardiac hypertro-
phy mediated through the up-regulation of PI3K/Akt and the
suppression of calcineurin/NF-AT3 signaling pathways in rats.
Life Sci 78(4):347–356
81. Westphal C etal (2012) Effects of estrogen, an ERα agonist and
raloxifene on pressure overload induced cardiac hypertrophy.
PLoS One7(12):e50802
82. Jazbutyte V etal (2008) Ligand-dependent activation of
ER{beta} lowers blood pressure and attenuates cardiac hyper-
trophy in ovariectomized spontaneously hypertensive rats.
Cardiovasc Res 77(4):774–781
83. Di Mattia RA etal (2020)The activation of the G protein-coupled
estrogen receptor (GPER) prevents and regresses cardiac hyper-
trophy. Life Sci242:117211
84. Wang H etal (2015) Activation of GPR30 inhibits cardiac
fibroblast proliferation. Mol Cell Biochem 405(1–2):135–148
85. Pei H etal (2019) G protein-coupled estrogen receptor 1 inhib-
its Angiotensin II-induced Cardiomyocyte Hypertrophy via the
regulation of PI3K-Akt-mTOR signalling and autophagy. Int J
Biol Sci 15(1):81–92
86. Verdin E etal (2010) Sirtuin regulation of mitochondria:
energy production, apoptosis, and signaling. Trends Biochem
Sci 35(12):669–675
87. Alcendor RR etal (2007) Sirt1 regulates aging and resistance
to oxidative stress in the heart. Circ Res 100(10):1512–1521
88. Palomer X etal (2013) An overview of the crosstalk between
inflammatory processes and metabolic dysregulation during dia-
betic cardiomyopathy. Int J Cardiol 168(4):3160–3172
89. Chang HC, Guarente L (2014) SIRT1 and other sirtuins in metab-
olism. Trends Endocrinol Metab 25(3):138–145
90. Tanno M et al (2010) Induction of manganese superoxide
dismutase by nuclear translocation and activation of SIRT1
promotes cell survival in chronic heart failure. J Biol Chem
285(11):8375–8382
91. Vinciguerra M etal (2009) Local IGF-1 isoform protects car-
diomyocytes from hypertrophic and oxidative stresses via SirT1
activity. Aging (Albany NY) 2(1):43–62
92. Hsu CP etal (2010) Silent information regulator 1 protects the
heart from ischemia/reperfusion. Circulation 122(21):2170–2182
93. Chen CJ etal (2009) Resveratrol protects cardiomyocytes from
hypoxia-induced apoptosis through the SIRT1-FoxO1 pathway.
Biochem Biophys Res Commun 378(3):389–393
94. Zhang QJ etal (2008) Endothelium-specific overexpres-
sion of class III deacetylase SIRT1 decreases atheroscle-
rosis in apolipoprotein E-deficient mice. Cardiovasc Res
80(2):191–199
95. Csiszar A etal (2009) Anti-oxidative and anti-inflammatory
vasoprotective effects of caloric restriction in aging: role of cir-
culating factors and SIRT1. Mech Ageing Dev 130(8):518–527
96. Vinciguerra M etal (2012) mIGF-1/JNK1/SirT1 signaling con-
fers protection against oxidative stress in the heart. Aging Cell
11(1):139–149
97. Furukawa A etal (2007) H2O2 accelerates cellular senes-
cence by accumulation of acetylated p53 via decrease in the
function of SIRT1 by NAD+ depletion. Cell Physiol Biochem
20(1–4):45–54
98. Arunachalam G etal (2010) SIRT1 regulates oxidant- and ciga-
rette smoke-induced eNOS acetylation in endothelial cells: Role
of resveratrol. Biochem Biophys Res Commun 393(1):66–72
99. Yao H, Rahman I (2012) Perspectives on translational and thera-
peutic aspects of SIRT1 in inflammaging and senescence. Bio-
chem Pharmacol 84(10):1332–1339
100. Planavila A etal (2011) Sirt1 acts in association with PPARα to
protect the heart from hypertrophy, metabolic dysregulation, and
inflammation. Cardiovasc Res 90(2):276–284
101. Gillum MP etal (2011) SirT1 regulates adipose tissue inflamma-
tion. Diabetes 60(12):3235–3245
102. Yang B etal (2018) Dioscin protects against coronary heart dis-
ease by reducing oxidative stress and inflammation via Sirt1/Nrf2
and p38 MAPK pathways. Mol Med Rep 18(1):973–980
103. Xie J etal (2020) Kallistatin alleviates heart failure in rats by
inhibiting myocardial inflammation and apoptosis via regulating
sirt1. Eur Rev Med Pharmacol Sci 24(11):6390–6399
104. Cohen HY etal (2004) Acetylation of the C terminus of Ku70
by CBP and PCAF controls Bax-mediated apoptosis. Mol Cell
13(5):627–638
105. Smith J (2002) Human Sir2 and the “silencing” of p53 activity.
Trends Cell Biol 12(9):404–406
106. Cheng HL etal (2003) Developmental defects and p53 hypera-
cetylation in Sir2 homolog (SIRT1)-deficient mice. Proc Natl
Acad Sci U S A 100(19):10794–10799
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Heart Failure Reviews
1 3
107. Alcendor RR etal (2004) Silent information regulator 2alpha, a
longevity factor and class III histone deacetylase, is an essential
endogenous apoptosis inhibitor in cardiac myocytes. Circ Res
95(10):971–980
108. Pillai JB etal (2005) Poly(ADP-ribose) polymerase-1-dependent
cardiac myocyte cell death during heart failure is mediated by
NAD+ depletion and reduced Sir2alpha deacetylase activity. J
Biol Chem 280(52):43121–43130
109. Hsu CP etal (2008) Sirt1 protects the heart from aging and stress.
Biol Chem 389(3):221–231
110. Han X etal (2020) Resveratrol protects H9c2 cells against
hypoxia-induced apoptosis through miR-30d-5p/SIRT1/NF-κB
axis. J Biosci45
111. Chong ZZ, Li F, Maiese K (2005) Stress in the brain: novel cel-
lular mechanisms of injury linked to Alzheimer’s disease. Brain
Res Brain Res Rev 49(1):1–21
112. Fontana L, Vinciguerra M, Longo VD (2012) Growth fac-
tors, nutrient signaling, and cardiovascular aging. Circ Res
110(8):1139–1150
113. Bolasco G etal (2012) Cardioprotective mIGF-1/SIRT1 signaling
induces hypertension, leukocytosis and fear response in mice.
Aging (Albany NY) 4(6):402–416
114. Harman D (1956) Aging: a theory based on free radical and
radiation chemistry. J Gerontol 11(3):298–300
115. Gracia-Sancho J etal (2010) Activation of SIRT1 by resveratrol
induces KLF2 expression conferring an endothelial vasoprotec-
tive phenotype. Cardiovasc Res 85(3):514–519
116. Salminen A, Kaarniranta K, Kauppinen A (2013) Crosstalk
between oxidative stress and SIRT1: impact on the aging process.
Int J Mol Sci 14(2):3834–3859
117. Xiong S etal (2011) FoxO1 mediates an autofeedback loop regu-
lating SIRT1 expression. J Biol Chem 286(7):5289–5299
118. Yamamoto T, Sadoshima J (2011) Protection of the heart against
ischemia/reperfusion by silent information regulator 1. Trends
Cardiovasc Med 21(1):27–32
119. Rajendran R etal (2011) Sirtuins: molecular traffic lights in the
crossroad of oxidative stress, chromatin remodeling, and tran-
scription. J Biomed Biotechnol2011:368276
120. Salminen A, Hyttinen JM, Kaarniranta K (2011) AMP-activated
protein kinase inhibits NF-κB signaling and inflammation: impact
on healthspan and lifespan. J Mol Med (Berl) 89(7):667–676
121. Xie QW, Kashiwabara Y, Nathan C (1994) Role of transcription
factor NF-kappa B/Rel in induction of nitric oxide synthase. J
Biol Chem 269(7):4705–4708
122. Mattagajasingh I etal (2007) SIRT1 promotes endothelium-
dependent vascular relaxation by activating endothelial nitric
oxide synthase. Proc Natl Acad Sci U S A 104(37):14855–14860
123. Potente M etal (2007) SIRT1 controls endothelial angiogenic
functions during vascular growth. Genes Dev 21(20):2644–2658
124. Sakamoto J etal (2004) Predominant expression of Sir2alpha,
an NAD-dependent histone deacetylase, in the embryonic mouse
heart and brain. FEBS Lett 556(1–3):281–286
125. Tanno M et al (2007) Nucleocytoplasmic shuttling of the
NAD+-dependent histone deacetylase SIRT1. J Biol Chem
282(9):6823–6832
126. Sundaresan NR etal (2011) The deacetylase SIRT1 promotes
membrane localization and activation of Akt and PDK1 during
tumorigenesis and cardiac hypertrophy. Sci Signal4(182):ra46
127. Sedding D, Haendeler J (2007) Do we age on Sirt1 expression?
Circ Res 100(10):1396–1398
128. Geng B etal (2013) PARP-2 knockdown protects cardiomyocytes
from hypertrophy via activation of SIRT1. Biochem Biophys Res
Commun 430(3):944–950
129. Wojciechowski P etal (2010) Resveratrol arrests and regresses
the development of pressure overload- but not volume overload-
induced cardiac hypertrophy in rats. J Nutr 140(5):962–968
130. Li S etal (2019) Fibroblast growth factor 21 protects the heart
from angiotensin II-induced cardiac hypertrophy and dys-
function via SIRT1. Biochim Biophys Acta Mol Basis Dis
1865(6):1241–1252
131. Dong HW, Zhang LF, Bao SL (2018) AMPK regulates
energy metabolism through the SIRT1 signaling pathway to
improve myocardial hypertrophy. Eur Rev Med Pharmacol Sci
22(9):2757–2766
132. Hou J etal (2010) Early apoptotic vascular signaling is deter-
mined by Sirt1 through nuclear shuttling, forkhead trafficking,
bad, and mitochondrial caspase activation. Curr Neurovasc Res
7(2):95–112
133. Hou J etal (2011) Erythropoietin employs cell longevity path-
ways of SIRT1 to foster endothelial vascular integrity during
oxidant stress. Curr Neurovasc Res 8(3):220–235
134. Margueron R etal (2004) Histone deacetylase inhibition and
estrogen signalling in human breast cancer cells. Biochem Phar-
macol 68(6):1239–1246
135. Reid G etal (2005) Multiple mechanisms induce transcriptional
silencing of a subset of genes, including oestrogen receptor
alpha, in response to deacetylase inhibition by valproic acid and
trichostatin A. Oncogene 24(31):4894–4907
136. Yao Y etal (2010) Inhibition of SIRT1 deacetylase suppresses
estrogen receptor signaling. Carcinogenesis 31(3):382–387
137. Liarte S, Alonso-Romero JL, Nicolás FJ (2018) SIRT1 and estro-
gen signaling cooperation for breast cancer onset and progres-
sion. Front Endocrinol (Lausanne) 9:552
138. Bendale DS etal (2013) 17-β Oestradiol prevents cardiovascular
dysfunction in post-menopausal metabolic syndrome by affecting
SIRT1/AMPK/H3 acetylation. Br J Pharmacol 170(4):779–795
139. Dyck JR, Lopaschuk GD (2006) AMPK alterations in cardiac
physiology and pathology: enemy or ally? J Physiol 574(Pt
1):95–112
140. Donato AJ etal (2011) SIRT-1 and vascular endothelial dys-
function with ageing in mice and humans. J Physiol 589(Pt
18):4545–4554
141. Meng Z etal (2016) Resveratrol attenuated estrogen-deficient-
induced cardiac dysfunction: role of AMPK, SIRT1, and mito-
chondrial function. Am J Transl Res 8(6):2641–2649
142. Guo JM etal (2017) SIRT1-dependent AMPK pathway in the
protection of estrogen against ischemic brain injury. CNS Neu-
rosci Ther 23(4):360–369
143. Elbaz A, Rivas D, Duque G (2009) Effect of estrogens on
bone marrow adipogenesis and Sirt1 in aging C57BL/6J mice.
Biogerontology 10(6):747–755
144. Rasbach KA, Schnellmann RG (2008) Isoflavones promote mito-
chondrial biogenesis. J Pharmacol Exp Ther 325(2):536–543
145. Elangovan S etal (2011) SIRT1 is essential for oncogenic signal-
ing by estrogen/estrogen receptor α in breast cancer. Cancer Res
71(21):6654–6664
146. Tao Z (2019)Estrogen signaling interacts with Sirt1 in adipocyte
autophagyVirginia Tech
147. Santolla MF etal (2015) SIRT1 is involved in oncogenic
signaling mediated by GPER in breast cancer. Cell Death Dis
6(7):e1834–e1834
Publisher's Note Springer Nature remains neutral with regard to
jurisdictional claims in published maps and institutional affiliations.
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
1.
2.
3.
4.
5.
6.
Terms and Conditions
Springer Nature journal content, brought to you courtesy of Springer Nature Customer Service Center GmbH (“Springer Nature”).
Springer Nature supports a reasonable amount of sharing of research papers by authors, subscribers and authorised users (“Users”), for small-
scale personal, non-commercial use provided that all copyright, trade and service marks and other proprietary notices are maintained. By
accessing, sharing, receiving or otherwise using the Springer Nature journal content you agree to these terms of use (“Terms”). For these
purposes, Springer Nature considers academic use (by researchers and students) to be non-commercial.
These Terms are supplementary and will apply in addition to any applicable website terms and conditions, a relevant site licence or a personal
subscription. These Terms will prevail over any conflict or ambiguity with regards to the relevant terms, a site licence or a personal subscription
(to the extent of the conflict or ambiguity only). For Creative Commons-licensed articles, the terms of the Creative Commons license used will
apply.
We collect and use personal data to provide access to the Springer Nature journal content. We may also use these personal data internally within
ResearchGate and Springer Nature and as agreed share it, in an anonymised way, for purposes of tracking, analysis and reporting. We will not
otherwise disclose your personal data outside the ResearchGate or the Springer Nature group of companies unless we have your permission as
detailed in the Privacy Policy.
While Users may use the Springer Nature journal content for small scale, personal non-commercial use, it is important to note that Users may
not:
use such content for the purpose of providing other users with access on a regular or large scale basis or as a means to circumvent access
control;
use such content where to do so would be considered a criminal or statutory offence in any jurisdiction, or gives rise to civil liability, or is
otherwise unlawful;
falsely or misleadingly imply or suggest endorsement, approval , sponsorship, or association unless explicitly agreed to by Springer Nature in
writing;
use bots or other automated methods to access the content or redirect messages
override any security feature or exclusionary protocol; or
share the content in order to create substitute for Springer Nature products or services or a systematic database of Springer Nature journal
content.
In line with the restriction against commercial use, Springer Nature does not permit the creation of a product or service that creates revenue,
royalties, rent or income from our content or its inclusion as part of a paid for service or for other commercial gain. Springer Nature journal
content cannot be used for inter-library loans and librarians may not upload Springer Nature journal content on a large scale into their, or any
other, institutional repository.
These terms of use are reviewed regularly and may be amended at any time. Springer Nature is not obligated to publish any information or
content on this website and may remove it or features or functionality at our sole discretion, at any time with or without notice. Springer Nature
may revoke this licence to you at any time and remove access to any copies of the Springer Nature journal content which have been saved.
To the fullest extent permitted by law, Springer Nature makes no warranties, representations or guarantees to Users, either express or implied
with respect to the Springer nature journal content and all parties disclaim and waive any implied warranties or warranties imposed by law,
including merchantability or fitness for any particular purpose.
Please note that these rights do not automatically extend to content, data or other material published by Springer Nature that may be licensed
from third parties.
If you would like to use or distribute our Springer Nature journal content to a wider audience or on a regular basis or in any other manner not
expressly permitted by these Terms, please contact Springer Nature at
onlineservice@springernature.com
... Previous studies have demonstrated that estrogen has a broad protective effect against cardiac diseases, such as dilated cardiomyopathy, cardiac hypertrophy and atherosclerosis (29)(30)(31). An epidemiological study has have demonstrated that premenopausal female patients have a lower risk of cardiovascular disease than male patients of the same age; however, after menopause, the risk in female patients is equal to or greater than that in male patients (32). ...
Article
Full-text available
17β-estradiol (E2) can inhibit cardiac fibrosis in female patients with heart failure (HF) and activate cell division cycle 42 (Cdc42), however it is unknown whether 17β-estradiol (E2) can ameliorate differentiation and collagen synthesis in TGF-β1-stimulated mouse cardiac fibroblasts (MCFs) by regulating cell division cycle 42 (Cdc42). The present study aimed to investigate the roles of estrogen and Cdc42 in preventing myocardial fibrosis and the underlying molecular mechanisms. An ELISA was used to measure the levels of E2 and Cdc42 in the serum of patients with heart failure (HF), and western blotting was used to measure the expression levels of Cdc42 in TGF-β1-stimulated immortalized MCFs. MCFs were transfected with a Cdc42 overexpression (OE) lentivirus or small interfering RNA (siRNA), or treated with a Cdc42 inhibitor (MLS-573151), and the function of Cdc42 was assessed by western blotting, immunofluorescence staining, reverse transcription-quantitative PCR and dual-luciferase reporter assays. Western blotting and immunofluorescence staining were performed to verify the protective effect of E2 on TGF-β1-stimulated MCFs, and the association between the protective effect and Cdc42. The results demonstrated that Cdc42 levels were increased in the serum of patients with HF and were positively correlated with the levels of E2; however, Cdc42 levels were decreased in TGF-β1-stimulated MCFs. Cdc42 inhibited MCF differentiation and collagen synthesis, as indicated by the protein expression of α-smooth muscle actin, collagen I and collagen III. Mechanistically, Cdc42 inhibited the transcription of TGF-β1 by promoting the expression of p21 (RAC1)-activated kinase 1 (Pak1)/JNK/c-Jun signaling pathway proteins and inhibiting the activity of the Tgfb1 gene promoter. In addition, E2 inhibited the differentiation and collagen synthesis of TGF-β1-stimulated MCFs, and promoted the protein expression of Pak1, JNK and c-Jun, consistent with the effects of Cdc42, whereas the effects of E2 were abolished when Cdc42 was knocked down. The aforementioned findings suggested that E2 could inhibit differentiation and collagen synthesis in TGF-β1-stimulated MCFs by regulating Cdc42 and the downstream Pak1/JNK/c-Jun signaling pathway.
... SIRT1 is a well-studied member of the sirtuin family and is known as a stress-responsive protein deacetylase [4]. Numerous studies have described the potential role of SIRT1 functions in DNA repair, oxidative stress response, metabolism, circadian control, and mitochondrial biogenesis [5][6][7]. Today, SIRT1 is targeted in many age-related human pathologies, such as cardiovascular disease, diabetes and other metabolic syndromes, chronic inflammation, neurodegenerative disorders, and cancer [8]. SIRT1 has a dual role in tumorigenesis, acting as both tumor suppressor and oncogene depending on the cancer type and stage [8]. ...
Article
Full-text available
Background Cancer cells are characterized by uncontrolled cell proliferation and impaired bioenergetics. Sirtuins are a family of highly conserved enzymes that play a fundamental role in energy metabolism regulation. SIRT1, in particular, drives many physiological stress responses and metabolic pathways following nutrient deprivation. We previously showed that SIRT1 activation using SCIC2.1 was able to attenuate genotoxic response and senescence. Here, we report that in hepatocellular carcinoma (HCC) cells under glucose-deprived conditions, SCIC2.1 treatment induced overexpression of SIRT1, SIRT3, and SIRT6, modulating metabolic response. Methods Flow cytometry was used to analyze the cell cycle. The MTT assay and xCELLigence system were used to measure cell viability and proliferation. In vitro enzymatic assays were carried out as directed by the manufacturer, and the absorbance was measured with an automated Infinite M1000 reader. Western blotting and immunoprecipitation were used to evaluate the expression of various proteins described in this study. The relative expression of genes was studied using real-time PCR. We employed a Seahorse XF24 Analyzer to determine the metabolic state of the cells. Oil Red O staining was used to measure lipid accumulation. Results SCIC2.1 significantly promoted mitochondrial biogenesis via the AMPK-p53-PGC1α pathway and enhanced mitochondrial ATP production under glucose deprivation. SIRT1 inhibition by Ex-527 further supported our hypothesis that metabolic effects are dependent on SIRT1 activation. Interestingly, SCIC2.1 reprogrammed glucose metabolism and fatty acid oxidation for bioenergetic circuits by repressing de novo lipogenesis. In addition, SCIC2.1-mediated SIRT1 activation strongly modulated antioxidant response through SIRT3 activation, and p53-dependent stress response via indirect recruitment of SIRT6. Conclusion Our results show that SCIC2.1 is able to promote energy homeostasis, attenuating metabolic stress under glucose deprivation via activation of SIRT1. These findings shed light on the metabolic action of SIRT1 in the pathogenesis of HCC and may help determine future therapies for this and, possibly, other metabolic diseases.
... Recent studies have identified a significant role for pathways of programmed cell death that can ultimately control cell survival and cognitive impairment during MS [46,61,128,130,132,134,[200][201][202] ( Table 1). Programmed cell death pathways that involve autophagy, apoptosis, pyroptosis, and ferroptosis can influence cell survival during inflammation [34,39,66,73,74,85,87,[203][204][205][206][207][208][209][210][211][212][213], oxidative stress [10,45,77,86,89,93,119,185,[214][215][216][217][218][219], ischemia [219][220][221][222][223], and mitochondrial dysfunction [32,83,85,115,198,[224][225][226][227][228][229][230][231][232] (Figure 1). Disorders of cellular metabolism that can lead to cognitive loss and other impairments, such as diabetes mellitus (DM), are also intimately tied to pathways of programmed cell death [24,72,115,208,214,216,219,224,[233][234][235][236][237][238][239][240][241]. ...
Article
Full-text available
Almost three million individuals suffer from multiple sclerosis (MS) throughout the world, a demyelinating disease in the nervous system with increased prevalence over the last five decades, and is now being recognized as one significant etiology of cognitive loss and dementia. Presently, disease modifying therapies can limit the rate of relapse and potentially reduce brain volume loss in patients with MS, but unfortunately cannot prevent disease progression or the onset of cognitive disability. Innovative strategies are therefore required to address areas of inflammation, immune cell activation, and cell survival that involve novel pathways of programmed cell death, mammalian forkhead transcription factors (FoxOs), the mechanistic target of rapamycin (mTOR), AMP activated protein kinase (AMPK), the silent mating type information regulation 2 homolog 1 (Saccharomyces cerevisiae) (SIRT1), and associated pathways with the apolipoprotein E (APOE-ε4) gene and severe acute respiratory syndrome coronavirus (SARS-CoV-2). These pathways are intertwined at multiple levels and can involve metabolic oversight with cellular metabolism dependent upon nicotinamide adenine dinucleotide (NAD+). Insight into the mechanisms of these pathways can provide new avenues of discovery for the therapeutic treatment of dementia and loss in cognition that occurs during MS.
Article
Full-text available
Purpose Alcohol is posited to affect sex steroid hormone concentrations, and a growing body of research has demonstrated menstrual cycle effects on women’s use of alcohol. The present targeted review synthesizes the literature examining the relationship between alcohol use and estradiol in women and suggests directions for future research. Methods Articles were identified using the PubMed database using the following criteria: published in English, presented original findings for women, were peerreviewed, and included measures of estradiol levels in the analyses. Twenty-nine articles were identified for inclusion. Results Results from this review indicate acute alcohol use temporarily increases estradiol levels in women, and this may be strongest when gonadotropins are high. Regular alcohol use (≥1 drink per day) increases estradiol levels, but estradiol appears to be suppressed in women with alcohol use disorders and physiologic dependence. Alcohol use tends to be highest in women during ovulation, when estradiol is high, and progesterone is low. Conclusion Alcohol use increases estradiol levels in women, particularly in the presence of gonadotropins. More research is needed to assess the effect of estradiol on alcohol use in women. Research on the relationship of estrogen and alcohol use in women is needed to elucidate health outcomes through the lifespan.
Article
Full-text available
Background and Objectives Long-term pathological myocardial hypertrophy (MH) seriously affects the normal function of the heart. Dronedarone was reported to attenuate left ventricular hypertrophy of mice. However, the molecular regulatory mechanism of dronedarone in MH is unclear. Methods Angiotensin II (Ang II) was used to induce cell hypertrophy of H9C2 cells. Transverse aortic constriction (TAC) surgery was performed to establish a rat model of MH. Cell size was evaluated using crystal violet staining and rhodamine phalloidin staining. Reverse transcription quantitative polymerase chain reaction and western blot were performed to detect the mRNA and protein expressions of genes. JASPAR and luciferase activity were conducted to predict and validate interaction between forkhead box O3 (FOXO3) and protein kinase inhibitor alpha (PKIA) promoter. Results Ang II treatment induced cell hypertrophy and inhibited sirtuin 1 (SIRT1) expression, which were reversed by dronedarone. SIRT1 overexpression or PKIA overexpression enhanced dronedarone-mediated suppression of cell hypertrophy in Ang II-induced H9C2 cells. Mechanistically, SIRT1 elevated FOXO3 expression through SIRT1-mediated deacetylation of FOXO3 and FOXO3 upregulated PKIA expression through interacting with PKIA promoter. Moreover, SIRT1 silencing compromised dronedarone-mediated suppression of cell hypertrophy, while PKIA upregulation abolished the influences of SIRT1 silencing. More importantly, dronedarone improved TAC surgery-induced MH and impairment of cardiac function of rats via affecting SIRT1/FOXO3/PKIA axis. Conclusions Dronedarone alleviated MH through mediating SIRT1/FOXO3/PKIA axis, which provide more evidences for dronedarone against MH.
Article
This study was designed to explore the protective effect and mechanism of naringin (NG) on radiation‐induced heart disease (RIHD) in rats. Rats were divided into four x‐ray (XR) irradiation groups with different absorbed doses (0/10/15/20 Gy), or into three groups (control, XR, and XR + NG groups). Subsequently, the ultrasonic diagnostic apparatus was adopted to assess and compare the left ventricular ejection fraction (LVEF), left ventricular fractional shortening (LVFS), left ventricular internal diameter at end diastole (LVIDd), and left ventricular internal diameter at end systole (LVIDs) in rats. Hematoxylin–eosin (H&E) staining and Masson staining were applied to detect the pathological damage and fibrosis of heart tissue. Western blot was used to measure the expression levels of myocardial fibrosis‐related proteins, endoplasmic reticulum stress‐related proteins, and Sirt1 (silent information regulator 1)/NF‐κB (nuclear factor kappa‐B) signaling pathway‐related proteins in cardiac tissues. Additionally, enzyme‐linked immunosorbent assay was utilized to detect the activities of pro‐inflammatory cytokines, malondialdehyde (MDA), superoxide dismutase (SOD), and catalase (CAT) in cardiac tissue. The results showed that NG treatment significantly attenuated the 20 Gy XR‐induced decline of LVEF and LVFS and the elevation of LVIDs. Cardiac tissue damage and fibrosis caused by 20 Gy XR were significant improved after NG treatment. Meanwhile, in rats irradiated by XR, marked downregulation was identified in the expressions of fibrosis‐related proteins (Col I, collagen type I; α‐SMA, α‐smooth muscle actin; and TGF‐β1, transforming growth factor‐beta 1) and endoplasmic reticulum stress‐related proteins (GRP78, glucose regulatory protein 78; CHOP, C/EBP homologous protein; ATF6, activating transcription factor 6; and caspase 12) after NG treatment. Moreover, NG treatment also inhibited the production of pro‐inflammatory cytokines [interleukin‐6, interleukin‐1β, and monocyte chemoattractant protein‐1 (MCP‐1)], reduced the expression of MDA, and promoted the activities of SOD and CAT. Also, NG treatment promoted Sirt1 expression and inhibited p65 phosphorylation. Collectively, XR irradiation induced cardiac injury in rats in a dose‐dependent manner. NG could improve the cardiac injury induced by XR irradiation by inhibiting endoplasmic reticulum stress and activating Sirt1/NF‐κB signaling pathway.
Article
Full-text available
Postmenopausal diabetic women are at higher risk to develop cardiovascular diseases (CVD) compared with nondiabetic women. Alterations in cardiac cellular metabolism caused by changes in sirtuins are one of the main causes of CVD in postmenopausal diabetic women. Several studies have demonstrated the beneficial actions of the G protein-coupled estrogen receptor (GPER) in postmenopausal diabetic CVD. However, the molecular mechanisms by which GPER has a cardioprotective effect are still not well understood. In this study, we used an ovariectomized (OVX) type-two diabetic (T2D) rat model induced by high-fat diet/streptozotocin to investigate the effect of G-1 (GPER-agonist) on sirtuins, and their downstream pathways involved in regulation of cardiac metabolism and function. Animals were divided into five groups: Sham-Control, T2D, OVX+T2D, OVX+T2D+Vehicle, and OVX+T2D+G-1. G-1 was administrated for six weeks. At the end, hemodynamic factors were measured, and protein levels of sirtuins, AMP-activated protein kinase (AMPK), and uncoupling protein 2 (UCP2) were determined by Western blot analysis. In addition, cardiac levels of oxidative stress biomarkers were measured. The findings showed that T2D led to left ventricular dysfunction and signs of oxidative stress in the myocardium, which were accompanied by decreased protein levels of Sirt1/2/3/6, p-AMPK, and UCP2 in the heart. Moreover, the induction of the menopausal state exacerbated these changes. In contrast, treatment with G-1 ameliorated the hemodynamic changes associated with ovariectomy by increasing Sirt1/3, p-AMPK, UCP2, and improving oxidative status. The results provide evidence of the cardioprotective effects of GPER operating through Sirt1/3, p-AMPK, and UCP2, thereby improving cardiac function. Our results suggest that increasing Sirt1/3 levels may offer new therapeutic approaches for postmenopausal diabetic CVD.
Article
Full-text available
Metabolic disorders and diabetes (DM) impact more than five hundred million individuals throughout the world and are insidious in onset, chronic in nature, and yield significant disability and death. Current therapies that address nutritional status, weight management, and pharmacological options may delay disability but cannot alter disease course or functional organ loss, such as dementia and degeneration of systemic bodily functions. Underlying these challenges are the onset of aging disorders associated with increased lifespan, telomere dysfunction, and oxidative stress generation that lead to multi-system dysfunction. These significant hurdles point to the urgent need to address underlying disease mechanisms with innovative applications. New treatment strategies involve non-coding RNA pathways with microRNAs (miRNAs) and circular ribonucleic acids (circRNAs), Wnt signaling, and Wnt1 inducible signaling pathway protein 1 (WISP1) that are dependent upon programmed cell death pathways, cellular metabolic pathways with AMP-activated protein kinase (AMPK) and nicotinamide, and growth factor applications. Non-coding RNAs, Wnt signaling, and AMPK are cornerstone mechanisms for overseeing complex metabolic pathways that offer innovative treatment avenues for metabolic disease and DM but will necessitate continued appreciation of the ability of each of these cellular mechanisms to independently and in unison influence clinical outcome.
Article
Full-text available
Life expectancy is increasing throughout the world and coincides with a rise in non-communicable diseases (NCDs), especially for metabolic disease that includes diabetes mellitus (DM) and neurodegenerative disorders. The debilitating effects of metabolic disorders influence the entire body and significantly affect the nervous system impacting greater than one billion people with disability in the peripheral nervous system as well as with cognitive loss, now the seventh leading cause of death worldwide. Metabolic disorders, such as DM, and neurologic disease remain a significant challenge for the treatment and care of individuals since present therapies may limit symptoms but do not halt overall disease progression. These clinical challenges to address the interplay between metabolic and neurodegenerative disorders warrant innovative strategies that can focus upon the underlying mechanisms of aging-related disorders, oxidative stress, cell senescence, and cell death. Programmed cell death pathways that involve autophagy, apoptosis, ferroptosis, and pyroptosis can play a critical role in metabolic and neurodegenerative disorders and oversee processes that include insulin resistance, β-cell function, mitochondrial integrity, reactive oxygen species release, and inflammatory cell activation. The silent mating type information regulation 2 homolog 1 (Saccharomyces cerevisiae) (SIRT1), AMP activated protein kinase (AMPK), and Wnt1 inducible signaling pathway protein 1 (WISP1) are novel targets that can oversee programmed cell death pathways tied to β-nicotinamide adenine dinucleotide (NAD⁺), nicotinamide, apolipoprotein E (APOE), severe acute respiratory syndrome (SARS-CoV-2) exposure with coronavirus disease 2019 (COVID-19), and trophic factors, such as erythropoietin (EPO). The pathways of programmed cell death, SIRT1, AMPK, and WISP1 offer exciting prospects for maintaining metabolic homeostasis and nervous system function that can be compromised during aging-related disorders and lead to cognitive impairment, but these pathways have dual roles in determining the ultimate fate of cells and organ systems that warrant thoughtful insight into complex autofeedback mechanisms.
Article
Full-text available
Acute kidney injury (AKI) is a common syndrome associated with high morbidity and mortality, despite progress in medical care. Many studies have shown that there are sex differences and different role of sex hormones particularly estrogens in kidney injury. In this regard, the incidence and rate of progression of kidney diseases are higher in men compared with women. These observations suggest that female sex hormone may be renoprotective. Silent information regulator 2 homolog 1 (SIRT1) is a histone deacetylase, which is implicated in multiple biologic processes in several organisms. In the kidneys, SIRT1 inhibits renal cell apoptosis, inflammation, and fibrosis. Studies have reported a link between SIRT1 and estrogen. In addition, SIRT1 regulates ERα expression and inhibition of SIRT1 activity suppresses ERα expression. This effect leads to inhibition of estrogen-responsive gene expression. In this text, we review the role of SIRT1 in mediating the protective effects of estrogen in the onset and progression of AKI.
Article
Full-text available
Objective: Heart failure (HF) is the loss of myocardial structure and function caused by various congenital or acquired heart diseases. This study explored the new target of treatment of HF by investigating the effect of Kallistatin (KS) on inflammation and apoptosis of myocardial tissue in HF rats. Materials and methods: We used doxorubicin to induce rat HF, and determined the success rate of modeling by detecting changes in rat heart weight and body weight, cardiac function and histology. We used two different doses (1 mg/kg, 2 mg/kg) of KS intraperitoneally injected rats and detected changes in inflammation and apoptosis of rat myocardial tissue by enzyme-linked immunosorbent assay (ELISA), Reverse Transcription-Polymerase Chain Reaction (RT-PCR) and immunohistochemical staining. Changes in the expression of sirt1 were also detected. In addition, we cultured rat myocardial cell line, H9c2 cells, and used siRNA-sirt1 to inhibit sirt1 in H9c2 cells to clarify the mechanism of KS regulating myocardial cells. Results: The body weight of HF rats treated with KS decreased while the heart weight increased. KS has also been found to reduce the concentration of brain natriuretic polypeptide (BNP) in rat serum. The results of echocardiography showed that KS effectively relieved the cardiac function of HF rats. Inflammatory factors (interleukin (IL)-1β, IL-6, IL-8 and tumor necrosis factor (TNF)-α) and pro-apoptotic molecules (caspase3/9 and Bax) in the serum and myocardial tissue of rats treated with KS were also significantly reduced. The inhibition of sirt1 in H9c2 cells significantly reduced the anti-apoptotic effect of KS on H9c2 cells. Conclusions: KS reduces the inflammation and apoptosis of myocardial tissue in HF rats by promoting the expression of sirt1, thereby alleviating HF-induced myocardial injury.
Article
Full-text available
Cardiovascular disease (CVD) is the number one cause of deaths worldwide, with yearly deaths due to atherothrombosis—i [...]
Article
Full-text available
Aging-induced progressive decline of molecular and metabolic factors in the myocardium is suggested to be related with heart dysfunction and cardiovascular disease. Therefore, we evaluated the effects of exercise training and l-arginine supplementation on oxidative stress, inflammation, and apoptosis in ventricle of the aging rat heart. Twenty-four 24-month-aged Wistar rats were randomly divided into four groups: the aged control, aged exercise, aged l-arginine (orally administered with 150 mg/kg for 12 weeks), and aged exercise + l-arginine groups. Six 4-month-old rats were also considered the young control. Animals with training program performed exercise on a treadmill 5 days/week for 12 weeks. After 12 weeks, protein levels of Bax, Bcl-2, pro-caspase-3/cleaved caspase-3, cytochrome C, and heat shock protein (HSP)-70 were assessed. Tissue contents of total anti-oxidant capacity, superoxide dismutase, catalase, and levels of tumor necrosis factor alpha (TNF-α), interleukin (IL)-1β, and IL-6 were analyzed. Histological and fibrotic changes were also evaluated. Treadmill exercise and l-arginine supplementation significantly alleviated aging-induced apoptosis with enhancing HSP-70 expression, increasing anti-oxidant enzyme activity, and suppressing inflammatory markers in the cardiac myocytes. Potent attenuation in apoptosis, inflammation, and oxidative stress was indicated in the rats with the combination of l-arginine supplementation and exercise program in comparison with each group (p < 0.05). In addition, fibrosis percentage and collagen accumulation were significantly lower in the rats with the combination treatment of l-arginine and exercise (p < 0.05). Treadmill exercise and l-arginine supplementation provided protection against age-induced increase in the myocyte loss and formation of fibrosis in the ventricle through potent suppression of oxidative stress, inflammations, and apoptosis pathways.
Article
Full-text available
The aging and elderly population are particularly susceptible to cardiovascular disease. Age is an independent risk factor for cardiovascular disease (CVD) in adults, but these risks are compounded by additional factors, including frailty, obesity, and diabetes. These factors are known to complicate and enhance cardiac risk factors that are associated with the onset of advanced age. Sex is another potential risk factor in aging adults, given that older females are reported to be at a greater risk for CVD than age-matched men. However, in both men and women, the risks associated with CVD increase with age, and these correspond to an overall decline in sex hormones, primarily of estrogen and testosterone. Despite this, hormone replacement therapies are largely shown to not improve outcomes in older patients and may also increase the risks of cardiac events in older adults. This review discusses current findings regarding the impacts of age and gender on heart disease.
Article
This 1956 paper describes a theory about mechanisms of aging that is based on free radical chemistry: "Aging and the degenerative diseases associated with it are attributed basically to the deleterious side attacks of free radicals on cell constituents and on the connective tissues. The free radicals probably arise largely through reactions involving molecular oxygen catalyzed in the cell by oxidative enzymes and in the connective tissues by traces of metals such as iron, cobalt, and manganese." Copyright (c) The Gerontological Society of America. Reproduced by permission of the publisher. Denham Harman, Aging: A Theory Based on Free Radical and Radiation Chemistry. J. Gerontol. 11 , 298-300 (1956).
Article
286 Michael Potente, Laleh Ghaeni, Molecular Cardiology, Frankfurt, Germany; Danila Baldessari, Zebrafish Group, Milan, Italy; Raul Mostoslavsky, Howard Hughes Medical Inst, The Children’s Hosp, Boston, MA; Lothar Rossig, Molecular Cardiology, Frankfurt, Germany; Franck Dequiedt, Cellular and Molecular Biology Unit, Gembloux, Belgium; Judith Haendeler, Molecular Cardiology, Frankfurt, Germany; Marina Mione, Zebrafish Group, Milan, Italy; Elisabetta Dejana, IFOM-FIRC Inst of Molecular Oncology, Milan, Italy; Frederick W Alt, Howard Hughes Medical Inst, The Children’s Hosp, Boston, MA; Andreas M Zeiher, Stefanie Dimmeler, Molecular Cardiology, Frankfurt, Germany Michael Potente, 2007 Finalist and Presenting Author
Article
The role of classical and non-classical estrogen receptors (ERs) in mediating the neuroprotective effects of this hormone on brain edema and long-term behavioral disorders was evaluated after traumatic brain injury (TBI). Ovariectomized rats were divided as follows: E2 (17 β-estradiol), E2-BSA (E2 conjugated to bovine serum albumin), G1 [G-protein-coupled estrogen receptor agonist (GPER)] or their vehicle was injected following TBI, whereas ICI (classical estrogen receptor antagonist), G15 (GPER antagonist), ICI + G15, and their vehicle were injected before the induction of TBI and the injection of E2 and E2-BSA. Brain water (BWC) and Evans blue (EB) contents were measured 24 h and 5 h after TBI, respectively. Intracranial pressure (ICP) and cerebral perfusion pressure (CPP) were measured before and at different times after TBI. Locomotor activity, anxiety-like behavior, and spatial memory were assessed on days 3, 7, 14, and 21 after injury. E2, E2-BSA, and G1 prevented the increase of BWC and EB content after TBI, and these effects were inhibited by ICI and G15. ICI and G15 also inhibited the beneficial effects of E2, E2-BSA on ICP, as well as CPP, after trauma. E2, E2-BSA, and G1 prevented the cognitive deficiency and behavioral abnormalities induced by TBI. Similar to the above parameters, ICI and G15 also reversed this E2 and E2-BSA effects on days 3, 7, 14, and 21. Our findings indicated that the beneficial effects of E2-BSA and E2 were inhibited by both ICI and G15, suggesting that GPER and classic ERs were involved in mediating the long-term effects of E2.
Article
Previous studies have demonstrated the cardioprotective role of resveratrol (Res). However, the underlying molecular mechanisms involved in the protective role of Res are still largely unknown. H9c2 cells were distributed into five groups: normal condition (Control), DMSO, 20 mMRes (dissolved with DMSO), hypoxia (Hyp), and Res+Hyp. Cell apoptosis was evaluated using flow cytometry and protein analysis of cleaved caspase 3 (cle-caspase 3). qRT-PCR assay was performed to measure the expression of microRNA-30d-5p (miR-30d-5p). MTT assay was performed to evaluate the cell proliferation. The relationship between miR-30d-5p and silent information regulator 1 (SIRT1) was confirmed by luciferase reporter, RNA immunoprecipitation (RIP), and western blot assays. Western blot was performed to analyze NF-κB/p65 and I-κBα expressions. Our data showed that hypoxia enhanced apoptosis andNF-κB signaling pathway, which was alleviated by Res treatment. Hypoxia increased the expression of miR-30d-5p while decreased the SIRT1expression, which was also attenuated by Res treatment. Furthermore, miR-30d-5p depletion inhibited the proliferation, reduced apoptosis and decreased the expression of cle-caspase 3 in H9c2 cells with hypoxia treatment. Luciferase reporter, RIP, and western blot assays further confirmed that miR-30d-5p negatively regulated the expression of SIRT1. Interestingly, the rescue-of-function experiments further indicated that knockdown of SIRT1 attenuated the effect of miR-30d-5p depletion on proliferation, apoptosis NF-κB signaling pathway inH9c2 cells with hypoxia treatment. In addition, the suppression of NF-κB signaling pathway increased cell viability while decreased cell apoptosis in hypoxia-mediatedH9c2 cells. Our data suggested Res mayprotectH9c2 cells against hypoxia-induced apoptosis through miR-30d-5p/SIRT1/NF-κB axis.