You are on page 1of 18

C H A P T E R

15
Role of mTOR Signaling in Cardioprotection
Anindita Das and Rakesh C. Kukreja
Pauley Heart Center, Division of Cardiology, Department of Internal Medicine, Virginia Commonwealth University
Medical Center, Richmond, VA, USA

15.1 INTRODUCTION 15.2 mTOR SIGNALING PATHWAY


The antifungal drug rapamycin (Sirolimuss) was mTOR belongs to the phosphatidylinositol 3-kinase
first isolated from the soil bacterium Streptomyces (PI3K)-related kinase family with a molecular weight of
hygroscopicus on Easter Island (Rapa Nui) in 1975 B289 kDa. It interacts with other molecular components
[1,2]. Subsequently, in the 1990s, the immunosup- to form two physically and functionally distinct com-
pressive property of rapamycin was recognized; it plexes, mTOR complex 1 (mTORC1) and mTOR complex
has since been primarily used in the treatment of 2 (mTORC2) [16]. Both complexes share the catalytic
organ rejection in transplant recipients [3 5]. The mTOR subunit, mLST8 (mammalian lethal with sec-13
antiproliferative effect of rapamycin, first discov- protein 8), Deptor (DEP domain-containing mTOR-inter-
ered in yeast system, is based on its ability to bind acting protein), and Tti1-Tel2 (Tel two interacting protein
to its intracellular receptor, the peptidyl-prolyl cis/ 1/Tel2) complex [17 19]. Other components of mTORC1
trans isomerase FKBP12 (FK506 binding protein) [6]. are Raptor (regulatory-associated protein of mTOR)
In the early 1990s, genetic screens in budding yeast and PRAS40 (proline-rich AKT substrate) [20,21].
identified TOR1 and TOR2 (target of rapamycin) as Alternatively, mTORC2 contains Rictor (rapamycin-insen-
mediators of antiproliferative effects of rapamycin sitive companion of mTOR) and mSin1 (mammalian
on yeast [7 9]. Shortly afterwards in 1994 and 1995, stress-activated MAP kinase-interacting protein 1) [22,23].
mammalian target of rapamycin (mTOR, also Raptor and Rictor function as scaffold proteins for
known as mechanistic TOR) was purified and char- integrity of the complexes; regulate their subcellular local-
acterized for the first time as the physical target of ization and interactions with substrates and regulator
rapamycin [10 12]. The rapamycin FKBP12 com- [24 26].
plex binds and specifically acts as an allosteric Over the past two decades, remarkable progress has
inhibitor of mTOR, an atypical serine/threonine been made to characterize the function and regulation
kinase [13]. Recently, a great deal of attention has of mTORC1, yet the mechanisms of mTORC2 activation
focused on the potential use of rapamycin for coat- are less well elucidated. mTORC1 is a signal integrator
ing drug-eluting stents to prevent restenosis follow- responding to multiple signals from growth factors,
ing coronary angioplasty [14]. Restenosis, a stressors such as hypoxia, nutrients, and energy status
reduction in lumen diameter after angioplasty and to control cellular homeostasis, growth, proliferation,
stent implantation, is the result of arterial damage and protein and lipid synthesis (Figure 15.1) [24,26].
with subsequent neointimal tissue proliferation. Due mTORC1 activates the ribosomal proteins S6 kinase 1
to its antiproliferative property, rapamycin prevents and 2 (S6K1/2) by phosphorylating their hydrophobic
intimal growth of graft coronary arteries and motif (HM), on Thr389 and Thr 388, respectively, which
reduces the incidence of vasculopathy [15]. promotes mRNA biogenesis, as well as translational

Molecules to Medicine with mTOR


DOI: http://dx.doi.org/10.1016/B978-0-12-802733-2.00003-7 245 © 2016 Elsevier Inc. All rights reserved.
246 15. ROLE OF mTOR SIGNALING IN CARDIOPROTECTION

Insulin or growth factors, cytokines, nutrients

IKKβ
Rag- PI3K
GTPase ERK1/2 p-IRS

Akt

TSC1/2
Rheb
Rapamycin mTORC2
mTORC1 Rictor
PRAS40 Raptor mSin1 Protor-1

PRAS40 mTOR mTOR Tti1/Tel2


Tti1/Tel2 Deptor
Deptor
mLST8
mLST8
PGC1α/
YY1
4EBP1 Akt PKCα RhoA
S6K1 ULK1/2- HIF1α
ATG SREBP1/2
Mitochondrial
biogenesis Growth Cytoskeletal
organization
Protein
synthesis Autophagy Lipogenesis Glycolysis Cell
survival Apoptosis

Cell
metabolism

FIGURE 15.1 The mammalian target of rapamycin (mTOR) signaling pathway. eIF4E-Bp1, eukaryotic translation initiation factor 4E-
binding protein 1; ATG, autophagy-related gene; DEPTOR, DEP domain-containing mTOR-interacting protein; ERK1/2, extracellular signal
regulated kinase 1/2; HIF-1α, hypoxia-inducible factor-1α; IKKβ, inhibitor of NF-κB kinase-β; p-IRS, phosphoinsulin receptor substrate;
mLST8, mammalian lethal with sec-13 protein 8; PGC1α, peroxisome proliferator-activated receptor γ coactivator-1α; PI3K, phosphoinositide
3-kinase; PKCα, protein kinase C α; PRAS40, proline-rich Akt substrate 40; Protor-1, protein observed with rictor-1; Raptor; regulatory-
associated protein in mTOR; Rheb, Ras homolog enriched in brain; RhoA, Ras homolog gene family, member A; S6K1, S6 kinase 1; SREBP1/2,
sterol regulatory element-binding protein 1/2; TSC1/2, tuberous sclerosis protein 1/2; Tt1, Tel two interacting protein 1; ULK1/2, unc-51-like
kinase 1/2; and YY1, Ying-Yang 1.

initiation and elongation of protein synthesis [25,27]. In and the translation of hypoxia-inducible factor 1α [30].
contrast, mTORC1 phosphorylates and inhibits eukary- Overall, mTORC1 has an evolutionarily conserved role
otic translation initiation factor 4E (eIF4E)-binding in promoting anabolic cell growth, but it also inhibits
protein 1 (4E-BP1), which triggers the initiation of cap- the catabolic process of autophagy, the central degrada-
dependent protein translation processes [25 27]. tion process of proteins and organelles in cells [24,31].
mTORC1 also activates the transcription factors sterol mTORC1 directly phosphorylates and suppresses
regulatory element-binding protein 1 and 2 (SREBP1/ ULK1/Atg13/FIP200 (unc-51-like kinase 1/mammalian
2), which control the expression of lipogenic genes and autophagy-related gene 13/focal adhesion kinase
promote the synthesis of cellular membrane lipids family-interacting protein of 200 kDa), a kinase complex
[25,28]. In addition, mTORC1 increases mitochondrial which promotes autophagosome formation [32,33].
DNA content and the expression of genes involved in mTORC1 also suppresses autophagy by regulating
oxidative metabolism through the peroxisome death-associated protein 1 and another crucial autopha-
proliferator-activated receptor γ (PPAR-γ)-mediated gic protein, Atg7 [34].
activation of transcription factor Ying-Yang 1, which The knowledge of mTORC2 regulation and function
ultimately promotes mitochondrial biogenesis and oxi- has lagged greatly behind that of mTORC1. mTORC2
dative metabolism [24,29]. Furthermore, mTORC1 signaling is insensitive to nutrient derivation, but it
increases glycolytic flux by activating the transcription responds to growth factors such as insulin via PI3

IV. mTOR IN CARDIOVASCULAR DEVELOPMENT AND DISEASE


15.3 mTOR IN MYOCARDIAL ISCHEMIA/REPERFUSION INJURY 247
kinase-dependent mechanisms (Figure 15.1) [35]. is a major cause of hundreds of thousands of deaths
mTORC2 phosphorylates the turn motif and the HM and disability each year worldwide. The most effective
of several AGC (protein kinase A, G, and C) kinases, therapeutic intervention for reducing acute myocardial
including PKB, SGK1 (serum/glucocorticoid-induced ischemic (AMI) injury by limiting the myocardial
kinase 1), PKCs (protein kinase C), and Rho1 infarct size as well as preserving left ventricular (LV)
(GDP GTP exchange protein-2 [Rom2]), resulting in function is early and effective myocardial reperfusion
their stabilization and activation [36]. Rictor enables [47]. Paradoxically, the reintroduction of oxygen-rich
mTORC2 to directly phosphorylate Akt at its HM blood during reperfusion to the ischemic tissue also has
Ser473 and facilitates Thr308 phosphorylation by detrimental effects, a phenomenon known as myocar-
PDK1 (phosphoinositide-dependent kinase 1) as part dial reperfusion injury [48,49]. Thus, the prevention of
of the insulin signaling cascade [37]. Akt regulates postischemic cardiac cell death may represent the best
essential cellular processes such as survival, growth, therapeutic modality to limit myocardial injury.
proliferation, metabolism, and apoptosis through the Although reperfusion is mandatory to salvage ische-
phosphorylation of several effectors [24]. mic myocardium from infarction [50,51], an irrevers-
Rictor of mTORC2 is directly phosphorylated by ible ischemic myocardial cell injury is developed after
S6K1 downstream of mTORC1 and this phosphoryla- prolonged coronary occlusion (after 2 h of ischemia)
tion event exerts a negative regulatory effect on the [52,53]. Therefore, in the last three decades, intensive
mTORC2-dependent phosphorylation of Akt-S473 research has focused on identifying the remarkable
within cells [38]. In contrast, upon stimulation with cardioprotective effect of innate adaptive responses to
growth factors, mTORC2 activates Akt which in turn ischemia/reperfusion (I/R) insult by different condi-
enhances mTORC1 activity through inactivation of tioning strategies which can provide therapeutic para-
TSC1/2 (tuberous sclerosis complex). The TSC2 is inac- digms for cardioprotection. The prime cardioprotective
tivated by Akt-dependent phosphorylation, which paradigm of ischemic preconditioning (IPC) was estab-
destabilizes TSC2 and disrupts its interaction with lished by Murry et al. in 1986 [54]. Protection using
TSC1 [39,40]. TSC1/2 complex inhibits mTORC1 IPC was accomplished by brief intermittent episodes
through its GTPase-activating protein activity toward of ischemia and reperfusion, resulting in substantial
Ras homolog enriched in brain (Rheb) [41]. These stud- myocardial protection from prolonged periods of
ies underscore the interconnection between mTORC1 ischemia. The identification of the mechanism of this
and mTORC2 [42]. Rapamycin preferentially inhibits phenomenon has provided a conceptual framework
mTORC1, however recent studies show that chronic for developing novel therapeutic strategies to mimick
treatment with the drug also inhibits mTORC2 [43,44]. the cardioprotecive effects of IPC with pharmacologi-
cal agents. IPC has been successfully translated to
humans with ischemic heart disease, but because of its
15.3 mTOR IN MYOCARDIAL ISCHEMIA/ nature (“pre”) can only be used in elective settings,
REPERFUSION INJURY such as percutaneous coronary interventions and coro-
nary artery bypass grafting, and not in AMI [55]. In
Cardiovascular disease (CVD) produces immense 2003, Zhao et al. identified another cardioprotective
health and economic burdens in the United States and phenomenon called postconditioning (PostC), which
globally. Despite advances in the treatment of CVD, it attenuates reperfusion injury [56]. PostC differs from
remains the leading global cause of death, accounting IPC in that it is instituted at the immediate onset of
for 17.3 million deaths per year, a number that is arterial reperfusion after a sustained episode of coro-
expected to grow to .23.6 million by 2030 [45]. CVDs nary occlusion. PostC has been successfully translated
are a group of disorders of the heart and blood vessels. to humans with ischemic heart disease, and as an
They include coronary heart disease, cerebrovascular intervention that is performed at immediate reperfu-
disease or heart stroke, heart attack (myocardial sion. Furthermore, it can also be used in patients
infarction), heart failure, dilated cardiomyopathy, undergoing interventional reperfusion of AMI [55].
hypertrophic cardiomyopathy, angina, arrhythmia, ath- Accumulating evidence indicates the important regu-
erosclerosis, hypertension, rheumatic heart disease, latory role of mTOR in myocardial I/R injury. Previous
peripheral vascular disease, congenital heart disease, studies report that ischemic [57,58] or pharmacological
and so on. Acute myocardial infarction (AMI), one of preconditioning/postconditioning [59,60] initiate PI3K-
the most catastrophic acute cardiac disorders, is the Akt signaling cascade and confer cardioprotection
major cause of the detrimental effects of coronary heart through recruitment of mTOR-p70S6K. In addition,
disease on the myocardium [46]. AMI, caused by acute mTOR inhibition with rapamycin abolished early [59]
myocardial ischemia due to insufficient coronary blood and delayed [58] preconditioning-induced cardioprotec-
flow to supply to meet the oxygen demand in the heart, tion as well as protection at reperfusion [57,60]. On the

IV. mTOR IN CARDIOVASCULAR DEVELOPMENT AND DISEASE


248 15. ROLE OF mTOR SIGNALING IN CARDIOPROTECTION

FIGURE 15.2 (A). Acute preconditioning with rapamycin (RAPA, 0.25 mg/kg, IP, administered 30 min before isolation of heart) reduces
infarct size in isolated mice hearts following 20 min global ischemia and 30 min reperfusion. Note that RAPA-induced reduction of infarct size is
blocked by 5-HD (5-hydroxydecanoate; 100 μM, infused 10 min before ischemia) ( P , 0.001 vs. DMSO control, DMSO 1 5-HD, RAPA 1 5-HD;
n 5 6 mice/group). (B, C) Effect of rapamycin on cardiomyocyte (B) necrosis and (C) apoptosis: Pretreatment with rapamycin for 1 h prior to
simulated ischemia (SI) for 40 min and reoxygenation (RO) for 1 or 18 h decreases the number of trypan blue-positive cells as well as TUNEL-
positive nuclei, indicating improved cell viability and reduced apoptosis with rapamycin (P, 0.001 vs. control; †P , 0.001 vs. SI-RO).

contrary, in 2006, the preconditioning-like cardiopro- Previous studies demonstrated that activation of JAK-
tective effect of rapamycin in isolated mouse heart and STAT signaling in response to IPC confers cardioprotec-
cardiomyocytes was first reported [61]. Rapamycin tion via prosurvival signaling cascades or inhibition of
treatment reduced infarct size after I/R injury by proapoptotic factors [64,65]. Rapamycin-induced cardio-
attenuating necrosis and apoptosis in cardiomyocytes protection against I/R injury was abolished with inhibi-
(Figure 15.2). This study also demonstrated that atten- tor of JAK2 (AG-490) or STAT3 (stattic) as well as in situ
uation of I/R injury with rapamycin was mediated knockdown of STAT3 [62] (Figure 15.4). Preconditioning
through opening of mitochondrial ATP-sensitive with rapamycin also induced phosphorylation of ERK,
potassium channels (mitoKATP channel). Later, the sig- STAT3-dependent eNOS, and glycogen synthase kinase-
naling pathways by which rapamycin triggers 3β (GSK-3β) in concert with increased prosurvival Bcl-2
preconditioning-like anti-infarct effect in ex vivo and to Bax ratio [62] (Figure 15.5).
in vivo myocardial I/R mouse models was further
investigated [62]. Rapamycin triggered unique cardio-
protective signaling through activation of Janus 15.4 ROLE OF mTORC1 VERSUS mTORC2
kinase 2 (JAK2)-STA3 [62] (Figure 15.3). The JAK- IN MYOCARDIAL INFARCTION
signal transducer and activator of transcription
(STAT) pathway is composed of a family of receptor- Several genetic and pharmacological approaches
associated cytosolic tyrosine kinases (JAKs) that phos- have now revealed that mTORC1 inhibition is benefi-
phorylate a tyrosine residue in cognate of STATs [63]. cial after myocardial infarction [66 69]. mTORC1 is

IV. mTOR IN CARDIOVASCULAR DEVELOPMENT AND DISEASE


15.4 ROLE OF mTORC1 VERSUS mTORC2 IN MYOCARDIAL INFARCTION 249

A B
DMSO RAPA
1.75

Ratio of p-STAT3/STAT3
1 2 3 4 5 1 2 3 4 5 1.50 *
p-STAT3 1.25
1.00
STAT3 0.75
0.50
Actin 0.25
0.00

SO

PA
A
M

R
D
FIGURE 15.3 Rapamycin (RAPA) enhances phosphorylation of STAT3 at tyrosine 705. (A) Representative immunoblots for p-STAT3, total
STAT3, and actin expression in whole heart after 1 h of RAPA treatment (0.25 mg/kg, IP). (B) Densitometry analysis of immunoblots for the
ratio of p-STAT3/STAT3.  P , 0.0001 vs. DMSO control, n 5 5 per group.

A Global I/R B In vivo I/R C


45 α α 50
40
40 *
α α
35 40 α 30
(% of risk area)

α
(% risk area)

30
Infarct size

Infarct size

FS (%)
25 30
20
20 20
15 *
10
10 * 10
5
0 0
0
tic
SO

SO

A
tic

tic
A
A

AP

AP
AP
0

tic

at
SO

tic
A

at

at
49

M
49
AP

at

St

St

St
at

+R
R
D

D
AG
M

St
AG

A+
St
R
D

tic
A+
A+

AP

at
AP
AP

St
R
R
R

D E

DMSO RAPA RAPA+Stattic Stattic


30 αβ
αβ
(% of total nuclei)
Apoptotic index

20

10

*
0
SO

tic

tic
AP

at

at
M

St

St
R
D

A+
AP
R

FIGURE 15.4 Inhibitors of JAK (AG-490) and STAT3 (Stattic) abolish infarct-limiting effect of rapamycin (RAPA) following I/R. Stattic
(20 mg/kg) or AG-490 (40 mg/kg) was administered intraperitoneally (IP) 30 min before rapamycin (0.25 mg/kg, IP) treatment. After 1 h of
rapamycin treatment, the heart is subjected to I/R. (A) Myocardial infarct size following 20 min of no-flow global ischemia and 30 min of
reperfusion,  P , 0.001 vs. DMSO (solvent of rapamycin); αP , 0.001 vs. RAPA, n 5 7 per group. (B) Myocardial infarct size following in situ I/
R by 30 min ligation of left coronary artery and 24 h reperfusion.  P , 0.001 vs. DMSO; αP , 0.01 vs. RAPA, n 5 6 per group. (C) Cardiac func-
tion (fractional shortening, FS), monitored by echocardiography following in situ I/R, is improved with rapamycin treatment. Stattic abolished
the rapamycin-induced functional improvement.  P , 0.05 vs. DMSO, n 5 5 per group. (D) Rapamycin reduces myocardial apoptosis, deter-
mined by TUNEL assay after in situ I/R. Stattic treatment blocks rapamycin-induced reduction of apoptosis. Representative images of
TUNEL-positive nuclei in green fluorescent color and total nuclei (blue) staining with 4,5-diamino-2-phenylindole (DAPI). (E) Bar diagram
showing quantitative data of TUNEL-positive nuclei in the peri-infarct region of myocardium.  P , 0.01 vs. DMSO; αP , 0.05 vs. RAPA;
β
P , 0.001 vs. DMSO; n 5 5 per group.

IV. mTOR IN CARDIOVASCULAR DEVELOPMENT AND DISEASE


250 15. ROLE OF mTOR SIGNALING IN CARDIOPROTECTION

and mTORC2 function in AAV-PRAS40-treated hearts.


Rapamycin
Conversely, mTORC2 deletion with AAV-sh-Rictor
exacerbated the decline in cardiac function and remo-
PD98059 ERK deling following a myocardial infarction. Such a combi-
natorial approach of inhibition of mTORC1 in
Stattic/
conjunction with activation of mTORC2 was important
STAT3
shSTAT3 for the regulation of ischemic damage and cardiac
?
remodeling after myocardial infarction. A previous
pGSK3β eNOS/iNOS
study also demonstrated the crucial role of mTORC2
Bcl-2 activation in myocardium protection from I/R injury
by constitutive phosphorylation of Akt-Ser473 in PH
MPTP MitoKATP
domain leucine-rich repeat protein phosphatase-1, a
selective Akt-Ser473 phosphatase, knockout mouse [72].
Mitochondrion
The pivotal role of mTORC2 has also been shown in
IPC [73]. In isolated perfused mouse hearts, IPC
induced mTORC2 activity, leading to phosphorylation
of Akt at Ser473 and subsequent phosphorylation of
Cardioprotection
GSK-3β, endothelial nitric oxide synthase, p70S6K, and
ribosomal S6. The protective effect of IPC was lost by
FIGURE 15.5 Proposed signaling pathways of STAT3-mediated pretreatment with dual mTORC inhibitors but not
cardioprotection by rapamycin against I/R injury.
with rapamycin, an mTORC1 inhibitor, which
confirmed the crucial role of mTORC2 activation in
cardioprotection [73].
activated in the remote myocardium during chronic mTOR overexpression was also found to reduce cell
myocardial infarction and contributes to ventricular death of ventricular cardiomyocytes in response to
remodeling [66,68]. Pharmacological mTOR inhibition hypoxia through activation of NF-κB and suppression
with everolimus prevents adverse LV remodeling and of inducible death factor BCL-2/adenovirus E1B
cardiac dilation, which limits infarct size with 19 kDa interacting protein 3 (Bnip3) [74]. Although, the
improved cardiac function following chronic myocar- relative activation of mTORC1 and C2 following over-
dial infarction [66]. The mTOR inhibition increased expression of mTOR was not examined in this study,
autophagy and decreased proteasome activity in the these findings raise the interesting possibility that acti-
border zone of the infarcted myocardium [66]. vation of Akt by mTORC2 may be required for direct
Rapamycin and S6K inhibitors alleviate cardiac ische- triggering of NF-κB signaling for averting cell death of
mic remodeling and cardiomyocyte apoptosis through ventricular myocytes during hypoxia [74,75].
PDK2 phosphorylation of Akt following myocardial Therefore, careful modulation of mTOR activity and a
infarction [70]. Since Rheb is required for mTORC1 more selective approach towards mTORC1 inhibition
signaling, a recent study reported that the partial may be critical for the treatment of myocardial
cardiac-specific deletion of Rheb1 and pharmacological ischemia.
inhibition of mTORC1 signaling exert cardioprotection Interestingly, the function of mTOR is distinct
against adverse cardiac remodeling in mice [71]. These between prolonged ischemia and reperfusion [76].
studies suggest that inhibition of mTORC1 could be a Since, mTOR is an essential regulator for protein syn-
potential therapeutic strategy to limit infarct size as thesis and cardiac hypertrophy, inhibition of mTOR as
well as attenuate adverse LV remodeling after myocar- well as protein synthesis during ischemia is beneficial
dial infarction. in reducing energy consumption and endoplasmic
Selective activation of mTORC2 with concurrent reticulum stress [76,77]. However, the cardioprotective
inhibition of mTORC1 signaling led to decreased cardi- role of mTOR signaling in reperfusion injury is still
omyocyte apoptosis and tissue damage after myocar- controversial. Earlier studies suggested that activation
dial infarction [68]. This study showed that selective of mTOR during the reperfusion phase may induce the
inhibition of mTORC1 with clinically relevant adeno- cell survival signaling mechanisms and mitochondrial
associated virus serotype 9 gene therapy with PRAS40 biogenesis, which may lead to recovery from myocar-
(AAV-PRAS40) signaling enhanced cardioprotection dial ischemia during reperfusion [29,60].
(by limiting myocardial infarction, improving cardiac Autophagy is an evolutionarily constitutive intracel-
function) and reduced mortality. Decreased mTORC1 lular catalytic process that targets dysfunctional or
signaling slowed degradation of insulin receptor damaged cytoplasmic constituents to the lysosome for
substrate-1, consequently improving insulin signaling degradation and recycling [78]. Autophagy is under

IV. mTOR IN CARDIOVASCULAR DEVELOPMENT AND DISEASE


15.5 mTOR IN CARDIAC HYPERTROPHY 251
the control of multiple signaling pathways, including contributes to cardiac dysfunction resulting in
nutrients, stress, hormones, growth factors, and intra- pressure-overloaded hearts of mice by enhanced cardi-
cellular energy information [79]. Autophagy was also omyocyte apoptosis through accumulation of proapop-
induced by ischemia and further enhanced by reperfu- totic proteins [93]. Akt is known to phosphorylate
sion in the mouse heart, in vivo [80]. Autophagy result- several proapoptotic substrates for subsequent UPS-
ing from ischemia was accompanied by activation of mediated elimination [93]. Rapamycin administration
AMPK and inhibited by dominant negative AMPK. augments activation of Akt, increases pressure
Importantly, autophagy plays distinct roles during overload-induced degradation of phosphorylated
ischemia and reperfusion: autophagy may be protec- inhibitor of κB, enhances expression of cellular inhibi-
tive during ischemia, whereas it may be detrimental tor of apoptosis protein 1, and decreases active
during reperfusion [80]. Since mTOR is another key caspase-3 [94]. Long-term rapamycin treatment
modulator of autophagy, the inhibition of GSK-3β and (2 weeks) in pressure-overload myocardium-blunted
resultant mTOR-dependent attenuation of autophagy hypertrophy, improved contractile function, and inhib-
are detrimental during prolonged ischemia, but they ited apoptosis by reducing caspase-3 and calpain acti-
are beneficial during reperfusion [76]. Moreover, exces- vation [94]. These studies indicate that an increase in
sive activation of autophagy during reperfusion leads targeted polyubiquitination (Ub), Akt activation, and
to cell death [80], although autophagy also plays a ben- survival signaling by inhibition of mTORC1 with rapa-
eficial role during myocardial reperfusion in some mycin during hypertrophy can tilt the molecular
experimental conditions [81]. Accumulating studies balance toward mTORC2 survival signaling in the
indicate that mTORC1 inhibition is protective during myocardium [94].
ischemia through the activation of autophagy, reduc- mTORC1 activity is strongly stimulated by upstream
tion of protein synthesis, and subsequent activation of Rheb in the heart and Rheb-mTORC1 signaling is
mTORC2 [42,69]. involved in pathologic heart remodeling [24]. A tar-
geted Rheb1-mTORC1 inhibition with astragaloside IV
or cardiac-specific rheb1 deletion effectively alleviated
15.5 mTOR IN CARDIAC HYPERTROPHY adverse cardiac remodeling after myocardial infarction
and hypertrophy in mouse models [71]. As previously
Cardiac hypertrophy, characterized by cardiomyo- underlined, among mTORC1 regulatory binding part-
cyte enlargement and structural remodeling, leads to ners, PRAS40 interacts with Raptor and inhibits Reb1-
myocardial wall thickness and is caused by physiologi- induced activation of mTORC1 kinase [21,95]. PRAS40
cal stimuli such as exercise, or pathological stimuli is phosphorylated by Akt in cardiomyocytes, which
from pressure or volume overload induced by hyper- relieves the inhibitory function of PRAS40 on mTORC1.
tension, myocardial infarction, or valvular heart dis- Cardiomyocyte-specific overexpression of PRAS40
ease [82]. Cardiac hypertrophy is, initially, an adaptive blocks mTORC1 in cardiomyocytes and blunts patho-
mechanism to maintain normal cardiac function. logical remodeling after long-term pressure overload
However, with prolonged non-reversible remodeling and preserves cardiac function [69].
this leads to dilated cardiomyopathy and heart failure However, other studies have challenged the impor-
[83]. This hypertrophic response is comprised of an tance of mTOR-mediated protein synthesis and growth
increase in cell size and protein content, as well as during hypertrophy. The cardiac hypertrophic growth
complex alterations in gene transcription and transla- in response to physiological and pathological stimuli
tion [84]. Since protein synthesis is regulated by the was not different in cardiac-specific transgenic mice
PI3K-AKT-mTOR signaling pathways, numerous stud- overexpressing mTOR or mutant mTOR compared
ies were conducted to understand the key role of with that of non-transgenic littermates [96,97].
mTOR in the development of cardiac hypertrophy. Interestingly, mTOR overexpression did not signifi-
Independent studies from different groups indicated cantly increase mTORC1 signaling during pathological
that the hypertrophic stimulus with β-adrenergic stim- hypertrophy induced by TAC in this study; instead, it
ulation [85], angiotensin-II [86], or insulin growth actually decreased phosphorylation of S6K [97].
factor-1 [87] induced mTORC1 activity in cardiomyo- Therefore, the specific activation of mTORC2 by
cytes, and the pharmacological inhibition of mTORC1 mTOR overexpression may contribute to preserve car-
with rapamycin reversed cardiac hypertrophy [88]. diac function during TAC. Moreover, the deletion of
Rapamycin also attenuated transverse aortic constric- the ribosomal S6 kinases, direct downstream effectors
tion (TAC)-induced cardiac hypertrophy as well as of mTOR, did not attenuate pathological, physiologi-
physical-exercise-induced physiological hypertrophy cal, or insulin-like growth factor receptor-induced
by inactivating mTORC1 [77,89 92]. Depression of cardiac hypertrophy [98]. Inducible cardiac-specific
ubiquitin proteasome system (UPS) activities raptor-deficient mice did not develop adaptive

IV. mTOR IN CARDIOVASCULAR DEVELOPMENT AND DISEASE


252 15. ROLE OF mTOR SIGNALING IN CARDIOPROTECTION

hypertrophy in response to pressure overload, (tuberous sclerosis complex 2) [39 41]. Rapamycin
however these mice developed ventricular dilation and blocks Akt-induced SREBP-1 expression and nuclear
cardiac dysfunction associated with autophagy, apo- accumulation, the expression of several lipogenic
ptosis, and mitochondrial and metabolic gene alter- genes, and the synthesis of various classes of lipids
ation [99]. These studies indicate that mTOR signaling [111]. The knockdown of Raptor, but not Rictor,
may be essential for adaptive cardiomyocyte growth to showed similar effects, indicating that SREBP-1 activa-
pressure overload. Further investigations are needed tion mainly depends on mTORC1, but not mTORC2
to better understand the specific role of mTORC1 and [28,112,113]. Using the mTORC1 inhibitor, rapamycin,
mTORC2 in cardiac hypertrophy. other independent studies also confirmed the signifi-
cant role of mTORC1 in the regulation of energy
production through profound effects on hepatic fatty
15.6 mTOR IN CARDIAC METABOLISM acid metabolism [114,115]. Additionally, the antidia-
AND DIABETES betic drug, metformin, which is a known to activate
adenosine monophosphate-activated protein kinase
The increasing prevalence of obesity and associated (AMPK) and also subsequently inhibits mTORC1,
metabolic disorders are well-recognized risk factors to reduced hepatic lipid content by promoting fatty acid
individual and public health in developing countries. oxidation, impairing SREBP-1c expression and cleav-
Metabolic syndrome (MS) is a constellation of obesity, age [116]. In addition, using liver-specific Rictor knock-
hypertension, and disorders of lipid and carbohydrate out mice, a recent study has established the crucial
metabolism. MS is a major risk factor for the develop- role of hepatic mTORC2 in lipogenesis through activa-
ment of type 2 diabetes, atherosclerosis, and coronary tion of Akt-mTORC1-SREBP-1c [117].
artery disease [100]. Since the mTOR signaling path- Pharmacological and genetic studies have demon-
way responds to nutrients and growth factor levels, its strated that mTORC1 signaling also plays a fundamen-
role in the regulation of metabolism has attracted tal role in lipid storage by stimulating the synthesis of
intense interest during the last decade, together with triglycerides in white adipose tissue (WAT) and the
the notion of intermittent versus persistent signaling to differentiation of preadipocytes into white adipocytes
explain its differential effects. A short-term activation through the translational control of the master regula-
of mTORC1 signaling during availability of nutrients tor of adipogenesis, PPAR-γ [118,119]. mTOR inhibi-
within a physiological range is necessary for anabolic tion with rapamycin reduced mRNA and protein
metabolism, energy storage, and consumption, as well levels of PPAR-γ and C/EBP-α (CCAAT/enhancer
as normal cell/tissue development [101]. This is binding protein) and the expression of numerous lipo-
supported by a study which reported that skeletal genic genes [118,120,121]. Constitutive activation of
muscle-specific Raptor-deficient mice begin to develop mTORC1 through TSC2 deletion or the deletion of 4E-
muscular dystrophy in conjunction with decreased BP1/2 induces PPAR-γ and C/EBP-α expression and
PGC-1α-mediated oxidative metabolism [102]. In promotes adipogenesis [102,119]. Additionally, S6K1-
contrast, a persistent activation of mTORC1 is involved deficient mice had reduced adipose tissue mass and
in the presence of nutritional excess including glucose were protected against diet-induced obesity [106].
and amino acids, genetic and diet-induced animal Adipocyte-specific deletion of Raptor in mice resulted
models of obesity, and metabolic disorders in the liver in lean mice with reduced WAT mass which are resis-
[103], skeletal muscle [103,104], adipose tissue tant to high-fat diet (HFD)-induced obesity [113].
[105,106], as well as in the heart [107,108].

15.8 mTOR IN DIABETIC HEARTS


15.7 mTOR SIGNALING IN ADIPOGENESIS
AND LIPOGENESIS MS, the combination of hypertension, obesity, dysli-
pidemia, and insulin resistance, is a precursor of dia-
mTORC1 activity is high in the livers of obese betes mellitus [122]. The diabetic heart shows early
rodents, which leads to degradation of IRS1 and poor maladaptation to alteration in lipid and/or glucose
insulin signaling, as well as hepatic insulin resistance metabolism, accompanied by lipid accumulation, and
[109]. Lipogenesis is paradoxically very active in the cardiac insulin resistance, which ultimately lead to
liver of insulin-resistant rodents [24]. mTORC1 hyper- cardiac dysfunction and heart failure [123,124].
activation by overfeeding promotes lipogenesis Hyperglycemia and hyperlipidemia as a result of dia-
through induction of SREBP-1c (SREBP) cleavage and betes lead to oxidative stress and inflammation in the
activation [110,111]. Insulin activates mTORC1 through cardiovascular system which causes endothelial dys-
Akt-mediated phosphorylation and inhibition of TSC2 function and cardiomyopathy [125,126]. Sustained

IV. mTOR IN CARDIOVASCULAR DEVELOPMENT AND DISEASE


15.8 mTOR IN DIABETIC HEARTS 253
hyperactive mTORC1 signaling plays a crucial role in contractile protein levels [134]. Autophagosome forma-
connecting metabolic stress and CVDs through stimu- tion and autophagic flux were also inhibited in the
lation of oxidative stress and inflammatory responses heart of the streptozotocin-induced type I mouse
[88,101]. Inhibition of AMPK during metabolic stress model [135]. Therefore, restoring myocardial cellular
leads to activation of mTORC1 [42,101,127]. Enhancing autophagy may represent a novel therapeutic target
AMPK activation during ischemia reduces infarct size for preserving myocardial structure and function in
in the diabetic heart following I/R injury size [128]. obesity and MS.
Moreover, the reduction of AMPK activity by ablation Recently, we reported the effect of chronic treatment
of liver kinase B1 (LKB1, a regulation of AMPK) in the (28 days) of rapamycin (0.25 mg/kg, intraperitoneally)
heart leads to energy deprivation and impairs cardiac in identifying novel protein targets involved in preser-
function during aerobic or ischemic conditions [129]. vation of cardiac function in type 2 diabetic mice [136].
In HFD-fed obese mice, increased activation of the The metabolic status was significantly improved in
Akt/mTOR pathway causes vascular senescence and these mice as shown by reductions in plasma glucose,
vascular dysfunction, which increase the susceptibility insulin, triglyceride levels, as well as body weight.
to peripheral and cerebral ischemia [130]. Rapamycin However, a similar treatment strategy with rapamycin
treatment of diet-induced obese mice or of transgenic did not affect body weight, metabolic parameters, or
mice with long-term activation of endothelial Akt cardiac function in non-diabetic mice. Another study
inhibited activation of mTOR and Akt, prevented vas- showed that long-term (6 months) treatment with
cular senescence without altering body weight, and rapamycin in female diabetic mice decreased body
reduced the severity of limb necrosis and ischemic weight and fat mass without affecting food intake. The
stroke [130]. In the HFD-induced obesity and MS markers of fatty acid oxidation and mitochondrial bio-
mouse model, enhanced cardiac mTORC1 activity sup- genesis were elevated in the gonadal WAT. In addi-
pressed autophagy and increased ischemic injury [67]. tion, circulating non-esterified free fatty acids were
Pharmacological and genetic inhibition of mTORC1 reduced, circulating adiponectin was elevated, and
restored autophagy and normalized the increase in insulin sensitivity was improved in these mice [137].
infarct size observed in HFD mice [67]. This study pro- Although earlier studies report that chronic treatment
vided a mechanistic explanation for the susceptibility with rapamycin (2 mg/kg/day) impairs whole-body
of ischemic heart with metabolic abnormality and insulin sensitivity by disrupting mTORC2 activity and
underscores the role of mTORC1 inhibition as an effec- blocking the ability of mTORC2-Akt to inhibit hepatic
tive therapeutic option to reduce cardiac ischemic gluconeogenesis [43,44]; a well-controlled cardiopro-
injury in the presence of MS. A reduction in myocar- tective dose of rapamycin (0.25 mg/kg/day) preferen-
dial autophagosome formation with associated induc- tially inhibited mTORC1, without disrupting mTORC2
tion of apoptosis inflammation, mitochondrial in type 2 diabetic mice [136]. Rapamycin inhibited the
dysfunction, and fibrosis are also reported in a swine enhanced phosphorylation of mTOR and S6 ribosomal
model of MS [131,132]. In another mouse model of protein (downstream target of mTORC1) due to diabe-
HFD-induced obesity, cardiac autophagosome forma- tes in the heart, without interfering with phosphoryla-
tion was reduced, with increased mTOR activity and tion of Akt (Ser473, target of mTORC2). A proteomic
cardiac hypertrophy and contractile dysfunction [133]. approach identified alteration of four cytoskeletal/con-
Rapamycin treatment restored cardiac function by pro- tractile proteins in diabetic mouse heart as compared
moting autophagy through inhibition of mTOR and to non-diabetic heart. The expression of the key con-
stimulation of AMPK activity [133]. Interestingly, HFD tractile/cytoskeletal proteins, MHC6α (myosin heavy
promoted the initiation of autophagy and accumula- polypeptide 6α) and MyBP-C (myosin binding protein
tion of autophagy, although it disrupted auto- C), were suppressed, but MLC-2 (myosin light chain 2)
phagosome maturation probably at the step of was increased in diabetic heart as compared to non-
autophagosome fusion with lysosomes, autophagic diabetic heart. Chronic treatment with rapamycin
flux [134]. As the major activator of mTOR, the Akt reversed the alteration of the cardiac expression of
family of serine threonine kinases is also activated by myosin isozymes.
HFD in the heart. HFD-feeding up-regulated cardiac Our proteomic study also identified the alteration of
expression of Akt2, but not Akt1 and Akt3. Akt2 three proteins involved in glucose metabolism [136].
knockout ameliorated HFD-feeding-induced cardiac These included Pgm2, glucose phosphomutase which
pathological hypertrophy and contractile anomalies by catalyzes the conversion of glucose 1 phosphate to the
facilitating cardiac autophagosome maturation pro- glycolytic intermediate glucose 6-phosphate.
cesses. Rescuing of the cardiac autophagy by Akt2 Suppression of Pgm2 in diabetic heart was completely
knockout inhibited apoptosis and improved mitochon- recovered following chronic rapamycin treatment
drial performance, intracellular Ca21 homeostasis, and [136]. PDH E1α, pyruvate dehydrogenase E1α,

IV. mTOR IN CARDIOVASCULAR DEVELOPMENT AND DISEASE


254 15. ROLE OF mTOR SIGNALING IN CARDIOPROTECTION

contributes to transforming pyruvate into acetyl-CoA,


Rapamycin
which can be used in the citric acid cycle to carry out
cellular respiration. Chronic inhibition of PDH with
the cardiac-specific overexpression of PDH inhibitor mTORC1
triggers an adaptive metabolic response in transgenic
mice [138]. The transgenic mice with overexpression of
cardiac-specific PDH are resistant to HFD-induced car- Endothelial dysfunction
diomyocyte lipid accumulation and exhibit normal
functional recovery after myocardial I/R injury. The Obesity Inflammation
adaptive metabolic reprogramming is involved with Insulin
resistance
the activation of AMPK kinase and induction of the
transcriptional coactivator, PGC-1α. Chronic treatment
Oxidative
with rapamycin reduced PDH E1α in diabetic heart, Hyperglycemia Type 2 diabetes
stress
with induction of PYGB protein (glycogen phosphory-
lase), which may improve cardiac metabolism and
help recover cardiac function [136]. Cardiac dysfunction
Increased production of reactive oxygen species
(ROS) in diabetic hearts is a major contributing factor
in the development and progression of diabetic cardio- Morbidity & mortality
myopathy [139,140]. In diabetic hearts, oxidative stress
is exacerbated in the presence of fatty acids, which FIGURE 15.6 Proposed scheme of cardioprotection by rapamycin
leads to mitochondrial uncoupling [140]. Therapeutic in type 2 diabetes. Rapamycin inhibits mTOR signaling and subse-
strategies that either reduce ROS production or aug- quently prevents endothelial dysfunction, obesity, hyperglycemia,
insulin resistance, inflammation, and oxidative stresses. This eventu-
ment myocardial antioxidant defense mechanisms
ally prevents diabetic-induced cardiac dysfunction in type 2
have been shown to be efficacious in the prevention of diabetes.
diabetes-induced myocardial dysfunction [141 144].
Interestingly, oxidative stress, as measured by glutathi-
mutant db/db mice [148]. PRAS40 overexpression pre-
one levels and lipid peroxidation, was significantly
vents cardiac dysfunction by blunting hypertrophic
reduced in rapamycin-treated diabetic hearts [136].
cardiac remodeling. In this context, a selective
Rapamycin treatment augmented the mitochondrial
approach towards mTORC1 inhibition may be a prom-
antioxidant enzyme PRX-5 (peroxiredoxin 5) in dia-
ising therapeutic modality in overcoming metabolic
betic hearts, which may help in alleviating oxidative
abnormalities and preventing cardiac dysfunction in
stress in the diabetic heart [136].
patients with diabetes.
Another powerful approach to overcome ROS-
induced toxicity is to limit the cellular availability of
transition metals, most notably iron sequestration [145].
An excess of free iron must be detoxified by sequester- 15.9 mTOR INHIBITION AND
ing in ferritin, the major intracellular iron storage pro- REPERFUSION THERAPY IN DIABETIC
tein, by oxidizing Fe(II) to Fe(III); ferritin heavy chain HEARTS
(FHC) possesses that ferroxidase activity [146].
A decrease in the abundance of FHC increases the The heart of a diabetic patient is more susceptible to
levels of iron deposition and oxidative stress, which AMI injury [122] and clinical studies suggest that diabe-
leads to cardiomyocyte death in failing hearts following tes is associated with higher mortality after AMI due to
ischemia or pressure overload [147]. The expression of more extensive atherosclerotic lesions, hypertrophy,
FHC is also induced with chronic rapamycin treatment and LV dysfunction [149,150]. Although reperfusion is
in the hearts of diabetic mice [136]. Therefore, it appears mandatory to salvage ischemic myocardium from
that chronic rapamycin treatment improves metabolic infarction, reperfusion itself contributes to irreversible
status and preserves cardiac function in diabetic mice, ischemic myocardial injury by formation of ROS, intra-
through attenuation of oxidative stress and alteration of cellular Ca21 overload, mitochondrial dysfunction, acti-
antioxidants as well as contractile and glucose meta- vation of intracellular proteolysis, and uncoordinated
bolic protein expression (Figure 15.6). excess contractile activity [49,55]. Currently there are no
A recent study shows that specific mTORC1 inhibi- therapies approved by the FDA that can directly dimin-
tion by PRAS40 prevents the development of diabetic ish the detrimental effect of myocardial reperfusion
cardiomyopathy by improving metabolic profile in injury. A recent, seminal study showed that rapamycin
HFD-induced type 2 diabetic mice and leptin receptor administered at the onset of reperfusion following

IV. mTOR IN CARDIOVASCULAR DEVELOPMENT AND DISEASE


15.10 CONCLUSIONS 255

A B

(n = 6)
(n = 4)
90

(% of pre-ischemia baseline)
80 (n = 4)

Rate-force product
C57 db/ db DMSO db / db RAPA 70
60
*
50
(n = 4)
40 40
*
(n = 4) 30
30 20
(% risk area)
Infarct size

10
(n = 6)
20 0
α

SO

PA
57
C

A
M

R
D
10

b
b

/d
/d

db
db
0
PA
SO
57
C

A
M

R
D

b
b

/d
/d

db
db

FIGURE 15.7 Reperfusion therapy with rapamycin (RAPA) reduces infarct size in diabetic mouse (db/db) hearts following ischemia/
reperfusion (I/R). (A) Myocardial infarct size in C57 and db/db mice following 30 min of no-flow global ischemia and 1 h of reperfusion in
Langendorff mode. Rapamycin (100 nM) or DMSO (solvent of rapamycin) is infused at the time of reperfusion ( P , 0.05 vs. C57; αP , 0.001
vs. other). (B) Product of heart rate and ventricular developed force (% of preischemic baseline) ( P , 0.05 vs. other).

global ischemia significantly reduces myocardial infarct significantly increased both total and phosphorylated
size in the hearts of diabetic mice [151] (Figure 15.7). STAT3 in the hearts of diabetic mice [151]
Moreover, rapamycin treatment improved cardiac func- (Figure 15.8). Rapamycin treatment at reperfusion
tion with increased coronary flow rates in diabetic heart exerted a robust infarct-sparing effect in HFD-fed
following I/R injury. Rapamycin treatment during wild-type mice which was associated with improved
reoxygenation following simulated ischemia also cardiac function after global I/R injury. Conversely,
reduced necrosis, apoptosis, and preserved mitochon- the infarct-limiting effect of rapamycin was abolished
drial membrane potential in adult primary cardiomyo- in HFD-fed cardiac-specific STAT3-deficient mice [151]
cytes isolated from diabetic mice [151]. Interestingly, (Figure 15.9). Moreover, the protective effect of rapa-
rapamycin infusion at the onset of reperfusion inhibited mycin treatment at reoxygenation in cardiomyocytes
phosphorylation of S6 ribosomal protein (target of isolated from HFD-fed STAT3-deficient mice was abol-
mTORC1) in hearts of normoglycemic mice without ished after SI/RO. These compelling studies revealed
interfering with Akt phosphorylation at Ser473, a target that reperfusion therapy with rapamycin in diabetic
of mTORC2 [151] (Figure 15.8). Contrary to expecta- hearts provides protection through the STAT3-AKT
tions, the phosphorylation of S6 was inhibited, but Akt signaling pathway.
phosphorylation was enhanced with rapamycin treat-
ment in diabetic hearts [151]. Thus specific inhibition of
mTORC1 activity as well as activation of mTORC2 may 15.10 CONCLUSIONS
provide beneficial effects of cardioprotection by rapa-
mycin treatment at reperfusion in diabetic heart follow- It is quite clear from the above-reviewed studies
ing I/R injury. that mTOR signaling is critical in cardiovascular and
The phosphorylation and subsequent activation of metabolic diseases. On the other hand, the clinical
STAT3 also contribute to cardioprotection via prosur- applications of mTOR inhibitors in these disorders
vival signaling cascade or the inhibition of proapopto- have not yet been established. Further studies are
tic factors after I/R injury [64,151]. The diabetic heart needed to clarify the cardioprotective mechanisms reg-
shows a decreased level of activated STAT3 which ulated by mTOR signaling in order to approve the clin-
is associated with insulin resistance and dilated ical use of mTOR inhibitors in patients. Thus, targeting
cardiomyopathy [151 153]. Rapamycin treatment mTOR signaling by rapamycin or its safer analogs

IV. mTOR IN CARDIOVASCULAR DEVELOPMENT AND DISEASE


A B n = 3, *P< 0.005 vs. other n = 3, *P< 0.01 vs. other
n = 3, *P< 0.05 vs. other
1.00 1.4 1.1
C57 db/db 1.2 * 1.0
0.9

p-STAT3/GAPDH
p-STAT3/STAT3
*

STAT3/GAPDH
0.75 1.0
* 0.8
0.7
DMSO RAPA DMSO RAPA 0.50
0.8
0.6
0.6 0.5
0.4
p-STAT3 0.4 0.3
0.25
(Tyr705) 0.2 0.2
0.1
0.00 0.0 0.0

SO

SO

A
SO

SO

A
SO

AP
STAT3

SO

AP
AP

AP
AP

M
AP

M
M

M
R

R
M

-R

-R
M

D
-D

-D
R

b-

b-
-R

b-

b-
-D

57

57
b-

57

57
/d

/d
b-

/d
57

/d
57

C
/d

db

db
C

C
/d

db

db
C

db
C

db
p-AKT
(S473)

n = 3, *P< 0.01 vs. C57-DMSO and C57-RAPA n = 3, *P< 0.05 vs. C57 and C57 RAPA
AKT α n = 3, *P< 0.05 vs. C57-RAPA αP< 0.001
P< 0.01 vs. db/db-DMSO vs. C57 and C57 RAPA
β
1.75 * 0.8 1.2
P< 0.01 vs. db/db
αβ
p-S6 1.50 *
(S235/236)
0.7 * * 1.0
*

p-AKT/GAPDH
1.25 0.6

p-AKT/AKT

AKT/GAPDH
0.8
1.00 0.5
0.4 0.6
S6 0.75
0.3 0.4
0.50
0.2
0.2
0.25 0.1
0.0
GAPDH 0.00 0.0

SO

A
SO

A
SO

A
SO

AP
SO

AP
SO

AP
AP

M
AP

M
AP

R
M

-R

D
M

-D
M

R
-R

b-
D
-D
R

b-
-R

57
b-
D
-D

57
b-

/d
57
b-

/d
57
b-

/d
57

C
/d

db
57

C
/d

db
C

db
/d

db
C

db
C

db
n = 3, *P< 0.001 vs. other
α
P< 0.01 vs. C57 n = 3, *P< 0.001 vs. other
n=3 α
P< 0.001 vs. C57
1.4
* 1.25
1.2 1.4
*
1.2
1.0 α 1.00
α α

p-S6/GAPDH
p-S6/S6

1.0

S6/GAPDH
0.8 0.75
0.8
0.6
0.50 0.6
0.4 0.4
0.25
0.2 0.2

0.0 0.00 0.0

SO

A
SO

A
SO

A
SO

A
SO

A
SO

AP
AP
AP
AP
AP
AP

M
M
M
M

R
-R
M

D
M

-D
-R

b-
-D
R
-R

b-
b-
D

57
-D

b-

57
57

/d
b-

57

/d
b-

/d
57

C
/d

db
57

db
/d

db
C
/d

db
C

db
C

db

FIGURE 15.8 Reperfusion therapy with rapamycin restores phosphorylation of STAT3 in diabetic heart, but inhibits mTORC1. Rapamycin (100 nM) or DMSO (solvent of rapamy-
cin) is infused at the time of reperfusion following 30 min of no-flow global ischemia in Langendorff mode. (A) Representative immunoblots of phospho-STAT3, STAT3, phospho-
AKT, AKT, phospho-S6, S6, and GAPDH in hearts of C57 and db/db mice. (B) Densitometric analysis of the ratios of phosphorylated (p) to total protein, total protein to GAPDH, and
phosphorylated proteins to GAPDH.
REFERENCES 257

A B
MCM NTG MCM TG
:STAT3 flox/flox :STAT3 flox/flox MCM.NTG:STAT3
Flox/Flox
MCM.TG:STAT3 Flox/Flox

STAT3 DMSO RAPA DMSO RAPA


Actin

*P< 0.03 vs. MCM NTG:STAT3flox/flox


1.5
STAT3/Actin

1.0

0.5
* n = 5; *P< 0.001 vs. other
45
0.0
MCM NTG: STAT3
flox/flox flox/flox 40
MCM TG: STAT3
35

(% risk area)
30

Infarct size
C n = 5; *P< 0.05 vs. other 25
α
P< 0.01 vs. other 20 *
(% of pre-ischemia baseline)

15
75 * 10
Rate-force product

5
50 0

PA
SO

SO
PA
α

A
M

M
A

R
D

D
R
25

MCM NTG:STAT flox/flox MCM TG:STAT flox/flox


0
PA

PA
SO

SO
A

A
M

M
R

R
D

MCM NTG:STAT flox/flox MCM TG:STAT flox/flox

FIGURE 15.9 Cardiac-specific STAT3 deficiency abolishes the infarct-limiting effect of rapamycin (RAPA) against ischemia/reperfusion
(I/R) injury in diabetic mice. (A) STAT3-deficient (MCM TG:STAT3flox/flox) and WT (MCM NTG:STAT3flox/flox) male mice (8 10 weeks) are
fed a high-fat diet (HFD) for 16 weeks, after which they receive tamoxifen (20 mg/kg/day IP) for 10 days. Rapamycin (100 nM) or DMSO (sol-
vent of rapamycin) is infused at the time of reperfusion following 30 min of no-flow global ischemia in Langendorff mode. Representative
immunoblots of STAT3 and GAPDH in hearts of HFD-fed and tamoxifen-treated STAT3-deficient and WT mice. Densitometric analysis repre-
senting fold change in STAT3/GAPDH ratio. (B) Myocardial infarct sizes of WT and STAT3-deficient mice following global I/R as well as
infusion of rapamycin (RAPA, 100 nM) or DMSO (solvent of rapamycin) at reperfusion. (C) Product of heart rate and ventricular developed
force (% of preischemic baseline).

with less adverse effects could be promising for treat- streptomycete and isolation of the active principle. J Antibiot
ment of cardiovascular and metabolic diseases includ- (Tokyo) 1975;28:721 6.
[3] Morris RE, Wu J, Shorthouse R. A study of the contrasting
ing I/R injury, hypertrophy, and type 2 diabetes. effects of cyclosporine, FK 506, and rapamycin on the suppres-
sion of allograft rejection. Transplant Proc 1990;22:1638 41.
Acknowledgments [4] Morris RE, Meiser BM, Wu J, Shorthouse R, Wang J. Use of rapa-
mycin for the suppression of alloimmune reactions in vivo:
This work was supported by grants from the National Institutes of schedule dependence, tolerance induction, synergy with cyclo-
Health R37 HL051045, R01 HL079424, R01 HL093685, and R01 sporine and FK 506, and effect on host-versus-graft and graft-
HL118808 (R. C. Kukreja) and A. D. Williams’ Fund of Virginia versus-host reactions. Transplant Proc 1991;23:521 4.
Commonwealth University Grant UL1RR031990 (A. Das) and CTSA [5] Stepkowski SM, Chen H, Daloze P, Kahan BD. Rapamycin, a potent
(UL1TR000058 from the National Center for Advancing Translational immunosuppressive drug for vascularized heart, kidney, and small
Sciences) and the CCTR (Center for Clinical and Translational bowel transplantation in the rat. Transplantation 1991;51:22 6.
Research) Endowment Fund (A. Das). [6] Koltin Y, Faucette L, Bergsma DJ, Levy MA, Cafferkey R, Koser
PL, et al. Rapamycin sensitivity in Saccharomyces cerevisiae is
mediated by a peptidyl-prolyl cis-trans isomerase related to
References human FK506-binding protein. Mol Cell Biol 1991;11:1718 23.
[1] Sehgal SN, Baker H, Vezina C. Rapamycin (AY-22,989), a new [7] Cafferkey R, Young PR, McLaughlin MM, Bergsma DJ, Koltin Y,
antifungal antibiotic. II. Fermentation, isolation and characteriza- Sathe GM, et al. Dominant missense mutations in a novel yeast
tion. J Antibiot (Tokyo) 1975;28:727 32. protein related to mammalian phosphatidylinositol 3-kinase and
[2] Vezina C, Kudelski A, Sehgal SN. Rapamycin (AY-22,989), a new VPS34 abrogate rapamycin cytotoxicity. Mol Cell Biol
antifungal antibiotic. I. Taxonomy of the producing 1993;13:6012 23.

IV. mTOR IN CARDIOVASCULAR DEVELOPMENT AND DISEASE


258 15. ROLE OF mTOR SIGNALING IN CARDIOPROTECTION

[8] Cafferkey R, McLaughlin MM, Young PR, Johnson RK, Livi GP. [26] Wullschleger S, Loewith R, Hall MN. TOR signaling in growth
Yeast TOR (DRR) proteins: amino-acid sequence alignment and and metabolism. Cell 2006;124:471 84.
identification of structural motifs. Gene 1994;141:133 6. [27] Ma XM, Blenis J. Molecular mechanisms of mTOR-mediated
[9] Kunz J, Henriquez R, Schneider U, Deuter-Reinhard M, Movva translational control. Nat Rev Mol Cell Biol 2009;10:307 18.
NR, Hall MN. Target of rapamycin in yeast, TOR2, is an essen- [28] Laplante M, Sabatini DM. An emerging role of mTOR in lipid
tial phosphatidylinositol kinase homolog required for G1 pro- biosynthesis. Curr Biol 2009;19:R1046 52.
gression. Cell 1993;73:585 96. [29] Cunningham JT, Rodgers JT, Arlow DH, Vazquez F, Mootha VK,
[10] Brown EJ, Albers MW, Shin TB, Ichikawa K, Keith CT, Lane Puigserver P. mTOR controls mitochondrial oxidative function
WS, et al. A mammalian protein targeted by G1-arresting rapa- through a YY1-PGC-1alpha transcriptional complex. Nature
mycin-receptor complex. Nature 1994;369:756 8. 2007;450:736 40.
[11] Sabatini DM, Erdjument-Bromage H, Lui M, Tempst P, Snyder SH. [30] Hudson CC, Liu M, Chiang GG, Otterness DM, Loomis DC,
RAFT1: a mammalian protein that binds to FKBP12 in a Kaper F, et al. Regulation of hypoxia-inducible factor 1 alpha
rapamycin-dependent fashion and is homologous to yeast TORs. expression and function by the mammalian target of rapamy-
Cell 1994;78:35 43. cin. Mol Cell Biol 2002;22:7004 14.
[12] Sabers CJ, Martin MM, Brunn GJ, Williams JM, Dumont FJ, [31] Sciarretta S, Zhai P, Volpe M, Sadoshima J. Pharmacological
Wiederrecht G, et al. Isolation of a protein target of the modulation of autophagy during cardiac stress. J Cardiovasc
FKBP12-rapamycin complex in mammalian cells. J Biol Chem Pharmacol 2012;60:235 41.
1995;270:815 22. [32] Egan DF, Shackelford DB, Mihaylova MM, Gelino S, Kohnz RA,
[13] Schmelzle T, Hall MN. TOR, a central controller of cell growth. MairW,etal.PhosphorylationofULK1(hATG1)byAMP-activated
Cell 2000;103:253 62. protein kinase connects energy sensing to mitophagy. Science
[14] Morice MC, Serruys PW, Sousa JE, Fajadet J, Ban HE, Perin M, 2011;331:456 61.
et al. A randomized comparison of a sirolimus-eluting stent [33] Ganley IG, Lam DH, Wang J, Ding X, Chen S, Jiang X. ULK1.
with a standard stent for coronary revascularization. N Engl J ATG13.FIP200 complex mediates mTOR signaling and is essen-
Med 2002;346:1773 80. tial for autophagy. J Biol Chem 2009;284:12297 305.
[15] Delgado JF, Manito N, Segovia J, Almenar L, Arizon JM, [34] Hsu PP, Kang SA, Rameseder J, Zhang Y, Ottina KA, Lim D,
Camprecios M, et al. The use of proliferation signal inhibitors et al. The mTOR-regulated phosphoproteome reveals a
in the prevention and treatment of allograft vasculopathy in mechanism of mTORC1-mediated inhibition of growth factor
heart transplantation. Transplant Rev (Orlando.) signaling. Science 2011;332:1317 22.
2009;23:69 79. [35] Gan X, Wang J, Su B, Wu D. Evidence for direct activation of
[16] Guertin DA, Sabatini DM. The pharmacology of mTOR inhibi- mTORC2 kinase activity by phosphatidylinositol 3,4,5-trispho-
tion. Sci Signal 2009;2:e24. sphate. J Biol Chem 2011;286:10998 1002.
[17] Kaizuka T, Hara T, Oshiro N, Kikkawa U, Yonezawa K, [36] Xie J, Herbert TP. The role of mammalian target of rapamycin
Takehana K, et al. Tti1 and Tel2 are critical factors in mamma- (mTOR) in the regulation of pancreatic beta-cell mass: implica-
lian target of rapamycin complex assembly. J Biol Chem tions in the development of type-2 diabetes. Cell Mol Life Sci
2010;285:20109 16. 2012;69:1289 304.
[18] Kim DH, Sarbassov DD, Ali SM, Latek RR, Guntur KV, [37] Sarbassov DD, Guertin DA, Ali SM, Sabatini DM.
Erdjument-Bromage H, et al. GbetaL, a positive regulator of the Phosphorylation and regulation of Akt/PKB by the rictor-
rapamycin-sensitive pathway required for the nutrient-sensitive mTOR complex. Science 2005;307:1098 101.
interaction between raptor and mTOR. Mol Cell [38] Dibble CC, Asara JM, Manning BD. Characterization of Rictor
2003;11:895 904. phosphorylation sites reveals direct regulation of mTOR com-
[19] Peterson TR, Laplante M, Thoreen CC, Sancak Y, Kang SA, plex 2 by S6K1. Mol Cell Biol 2009;29:5657 70.
Kuehl WM, et al. DEPTOR is an mTOR inhibitor frequently [39] Inoki K, Li Y, Zhu T, Wu J, Guan KL. TSC2 is phosphorylated
overexpressed in multiple myeloma cells and required for their and inhibited by Akt and suppresses mTOR signalling. Nat
survival. Cell 2009;137:873 86. Cell Biol 2002;4:648 57.
[20] Kim DH, Sarbassov DD, Ali SM, King JE, Latek RR, Erdjument- [40] Manning BD, Tee AR, Logsdon MN, Blenis J, Cantley LC.
Bromage H, et al. mTOR interacts with raptor to form a Identification of the tuberous sclerosis complex-2 tumor sup-
nutrient-sensitive complex that signals to the cell growth pressor gene product tuberin as a target of the phosphoinosi-
machinery. Cell 2002;110:163 75. tide 3-kinase/akt pathway. Mol Cell 2002;10:151 62.
[21] Sancak Y, Thoreen CC, Peterson TR, Lindquist RA, Kang SA, [41] Dibble CC, Elis W, Menon S, Qin W, Klekota J, Asara JM, et al.
Spooner E, et al. PRAS40 is an insulin-regulated inhibitor of the TBC1D7 is a third subunit of the TSC1-TSC2 complex upstream
mTORC1 protein kinase. Mol Cell 2007;25:903 15. of mTORC1. Mol Cell 2012;47:535 46.
[22] Jacinto E, Facchinetti V, Liu D, Soto N, Wei S, Jung SY, et al. [42] Sciarretta S, Volpe M, Sadoshima J. Mammalian target of rapa-
SIN1/MIP1 maintains rictor-mTOR complex integrity and regu- mycin signaling in cardiac physiology and disease. Circ Res
lates Akt phosphorylation and substrate specificity. Cell 2014;114:549 64.
2006;127:125 37. [43] Lamming DW, Ye L, Katajisto P, Goncalves MD, Saitoh M,
[23] Sarbassov DD, Ali SM, Kim DH, Guertin DA, Latek RR, Stevens DM, et al. Rapamycin-induced insulin resistance is
Erdjument-Bromage H, et al. Rictor, a novel binding partner of mediated by mTORC2 loss and uncoupled from longevity.
mTOR, defines a rapamycin-insensitive and raptor-independent Science 2012;335:1638 43.
pathway that regulates the cytoskeleton. Curr Biol [44] Sarbassov DD, Ali SM, Sengupta S, Sheen JH, Hsu PP, Bagley
2004;14:1296 302. AF, et al. Prolonged rapamycin treatment inhibits mTORC2
[24] Laplante M, Sabatini DM. mTOR signaling in growth control assembly and Akt/PKB. Mol Cell 2006;22:159 68.
and disease. Cell 2012;149:274 93. [45] Mozaffarian D, Benjamin EJ, Go AS, Arnett DK, Blaha MJ,
[25] Laplante M, Sabatini DM. Regulation of mTORC1 and its Cushman M, et al. Heart disease and stroke statistics—2015
impact on gene expression at a glance. J Cell Sci update: a report from the American Heart Association.
2013;126:1713 19. Circulation 2015;131:e29 322.

IV. mTOR IN CARDIOVASCULAR DEVELOPMENT AND DISEASE


REFERENCES 259
[46] Hausenloy DJ, Yellon DM. Myocardial ischemia-reperfusion [65] Xuan YT, Guo Y, Han H, Zhu Y, Bolli R. An essential role of
injury: a neglected therapeutic target. J Clin Invest the JAK-STAT pathway in ischemic preconditioning. Proc Natl
2013;123:92 100. Acad Sci U S A 2001;98:9050 5.
[47] Hochman JS, Choo H. Limitation of myocardial infarct expan- [66] Buss SJ, Muenz S, Riffel JH, Malekar P, Hagenmueller M, Weiss
sion by reperfusion independent of myocardial salvage. CS, et al. Beneficial effects of mammalian target of rapamycin
Circulation 1987;75:299 306. inhibition on left ventricular remodeling after myocardial
[48] Braunwald E, Kloner RA. Myocardial reperfusion: a double- infarction. J Am Coll Cardiol 2009;54:2435 46.
edged sword? J Clin Invest 1985;76:1713 19. [67] Sciarretta S, Zhai P, Shao D, Maejima Y, Robbins J, Volpe M,
[49] Yellon DM, Hausenloy DJ. Myocardial reperfusion injury. N et al. Rheb is a critical regulator of autophagy during myocar-
Engl J Med 2007;357:1121 35. dial ischemia: pathophysiological implications in obesity and
[50] Ginks WR, Sybers HD, Maroko PR, Covell JW, Sobel BE, Ross metabolic syndrome. Circulation 2012;125:1134 46.
Jr J. Coronary artery reperfusion. II. Reduction of myocardial [68] Volkers M, Konstandin MH, Doroudgar S, Toko H, Quijada P,
infarct size at 1 week after the coronary occlusion. J Clin Invest Din S, et al. Mechanistic target of rapamycin complex 2 pro-
1972;51:2717 23. tects the heart from ischemic damage. Circulation
[51] Maroko PR, Libby P, Ginks WR, Bloor CM, Shell WE, Sobel BE, 2013;128:2132 44.
et al. Coronary artery reperfusion. I. Early effects on local myo- [69] Volkers M, Toko H, Doroudgar S, Din S, Quijada P, Joyo AY,
cardial function and the extent of myocardial necrosis. J Clin et al. Pathological hypertrophy amelioration by PRAS40-
Invest 1972;51:2710 16. mediated inhibition of mTORC1. Proc Natl Acad Sci USA
[52] Reimer KA, Lowe JE, Rasmussen MM, Jennings RB. The wave- 2013;110:12661 6.
front phenomenon of ischemic cell death. 1. Myocardial infarct [70] Di R, Wu X, Chang Z, Zhao X, Feng Q, Lu S, et al. S6K inhibi-
size vs duration of coronary occlusion in dogs. Circulation tion renders cardiac protection against myocardial infarction
1977;56:786 94. through PDK1 phosphorylation of Akt. Biochem J
[53] Reimer KA, Jennings RB. The “wavefront phenomenon” of 2012;441:199 207.
myocardial ischemic cell death. II. Transmural progression of [71] Wu X, Cao Y, Nie J, Liu H, Lu S, Hu X, et al. Genetic and phar-
necrosis within the framework of ischemic bed size (myocar- macological inhibition of Rheb1-mTORC1 signaling exerts car-
dium at risk) and collateral flow. Lab Invest 1979;40:633 44. dioprotection against adverse cardiac remodeling in mice. Am J
[54] Murry CE, Jennings RB, Reimer KA. Preconditioning with Pathol 2013;182:2005 14.
ischemia: a delay of lethal cell injury in ischemic myocardium. [72] Miyamoto S, Purcell NH, Smith JM, Gao T, Whittaker R, Huang K,
Circulation 1986;74:1124 36. et al. PHLPP-1 negatively regulates Akt activity and survival in
[55] Heusch G. Molecular basis of cardioprotection: signal transduc- the heart. Circ Res 2010;107:476 84.
tion in ischemic pre-, post-, and remote conditioning. Circ Res [73] Yano T, Ferlito M, Aponte A, Kuno A, Miura T, Murphy E,
2015;116:674 99. et al. Pivotal role of mTORC2 and involvement of ribosomal
[56] Zhao ZQ, Corvera JS, Halkos ME, Kerendi F, Wang NP, protein S6 in cardioprotective signaling. Circ Res
Guyton RA, et al. Inhibition of myocardial injury by ischemic 2014;114:1268 80.
postconditioning during reperfusion: comparison with ischemic [74] Dhingra R, Gang H, Wang Y, Biala AK, Aviv Y, Margulets V,
preconditioning. Am J Physiol Heart Circ Physiol 2003;285: et al. Bidirectional regulation of nuclear factor-kappaB and
H579 88. mammalian target of rapamycin signaling functionally links
[57] Hausenloy DJ, Mocanu MM, Yellon DM. Cross-talk between Bnip3 gene repression and cell survival of ventricular myo-
the survival kinases during early reperfusion: its contribution cytes. Circ Heart Fail 2013;6:335 43.
to ischemic preconditioning. Cardiovasc Res 2004;63:305 12. [75] Regula KM, Baetz D, Kirshenbaum LA. Nuclear factor-kappaB
[58] Kis A, Yellon DM, Baxter GF. Second window of protection fol- represses hypoxia-induced mitochondrial defects and cell death
lowing myocardial preconditioning: an essential role for PI3 of ventricular myocytes. Circulation 2004;110:3795 802.
kinase and p70S6 kinase. J Mol Cell Cardiol 2003;35:1063 71. [76] Zhai P, Sciarretta S, Galeotti J, Volpe M, Sadoshima J.
[59] Gross ER, Hsu AK, Gross GJ. Opioid-induced cardioprotection Differential roles of GSK-3beta during myocardial ischemia and
occurs via glycogen synthase kinase beta inhibition during ischemia/reperfusion. Circ Res 2011;109:502 11.
reperfusion in intact rat hearts. Circ Res 2004;94:960 6. [77] McMullen JR, Sherwood MC, Tarnavski O, Zhang L, Dorfman
[60] Jonassen AK, Sack MN, Mjos OD, Yellon DM. Myocardial pro- AL, Shioi T, et al. Inhibition of mTOR signaling with rapamycin
tection by insulin at reperfusion requires early administration regresses established cardiac hypertrophy induced by pressure
and is mediated via Akt and p70s6 kinase cell-survival signal- overload. Circulation 2004;109:3050 5.
ing. Circ Res 2001;89:1191 8. [78] Mizushima N, Levine B, Cuervo AM, Klionsky DJ. Autophagy
[61] Khan S, Salloum F, Das A, Xi L, Vetrovec GW, Kukreja RC. fights disease through cellular self-digestion. Nature
Rapamycin confers preconditioning-like protection against 2008;451:1069 75.
ischemia-reperfusion injury in isolated mouse heart and cardio- [79] Glick D, Barth S, Macleod KF. Autophagy: cellular and molecu-
myocytes. J Mol Cell Cardiol 2006;41:256 64. lar mechanisms. J Pathol 2010;221:3 12.
[62] Das A, Salloum FN, Durrant D, Ockaili R, Kukreja RC. [80] Matsui Y, Takagi H, Qu X, Abdellatif M, Sakoda H, Asano T,
Rapamycin protects against myocardial ischemia-reperfusion et al. Distinct roles of autophagy in the heart during ischemia
injury through JAK2-STAT3 signaling pathway. J Mol Cell and reperfusion: roles of AMP-activated protein kinase and
Cardiol 2012;53:858 69. Beclin 1 in mediating autophagy. Circ Res 2007;100:914 22.
[63] Dawn B, Xuan YT, Guo Y, Rezazadeh A, Stein AB, Hunt G, [81] Hamacher-Brady A, Brady NR, Gottlieb RA. The interplay
et al. IL-6 plays an obligatory role in late preconditioning via between pro-death and pro-survival signaling pathways in
JAK-STAT signaling and upregulation of iNOS and COX-2. myocardial ischemia/reperfusion injury: apoptosis meets
Cardiovasc Res 2004;64:61 71. autophagy. Cardiovasc Drugs Ther 2006;20:445 62.
[64] Suleman N, Somers S, Smith R, Opie LH, Lecour SC. Dual acti- [82] Heineke J, Molkentin JD. Regulation of cardiac hypertrophy by
vation of STAT-3 and Akt is required during the trigger phase intracellular signalling pathways. Nat Rev Mol Cell Biol
of ischaemic preconditioning. Cardiovasc Res 2008;79:127 33. 2006;7:589 600.

IV. mTOR IN CARDIOVASCULAR DEVELOPMENT AND DISEASE


260 15. ROLE OF mTOR SIGNALING IN CARDIOPROTECTION

[83] Nadal-Ginard B, Kajstura J, Leri A, Anversa P. Myocyte death, hypertrophy, alters metabolic gene expression, and causes
growth, and regeneration in cardiac hypertrophy and failure. heart failure in mice. Circulation 2011;123:1073 82.
Circ Res 2003;92:139 50. [100] Eberly LE, Prineas R, Cohen JD, Vazquez G, Zhi X, Neaton JD,
[84] Dorn GW, Force T. Protein kinase cascades in the regulation et al. Metabolic syndrome: risk factor distribution and 18-year
of cardiac hypertrophy. J Clin Invest 2005;115:527 37. mortality in the multiple risk factor intervention trial. Diabetes
[85] Simm A, Schluter K, Diez C, Piper HM, Hoppe J. Activation of Care 2006;29:123 30.
p70(S6) kinase by beta-adrenoceptor agonists on adult cardio- [101] Yang Z, Ming XF. mTOR signalling: the molecular interface
myocytes. J Mol Cell Cardiol 1998;30:2059 67. connecting metabolic stress, aging and cardiovascular dis-
[86] Sadoshima J, Izumo S. Rapamycin selectively inhibits angio- eases. Obes Rev 2012;13(Suppl. 2):58 68.
tensin II-induced increase in protein synthesis in cardiac myo- [102] Bentzinger CF, Romanino K, Cloetta D, Lin S, Mascarenhas JB,
cytes in vitro. Potential role of 70-kD S6 kinase in angiotensin Oliveri F, et al. Skeletal muscle-specific ablation of raptor, but
II-induced cardiac hypertrophy. Circ Res 1995;77:1040 52. not of rictor, causes metabolic changes and results in muscle
[87] Lavandero S, Foncea R, Perez V, Sapag-Hagar M. Effect of dystrophy. Cell Metab 2008;8:411 24.
inhibitors of signal transduction on IGF-1-induced protein syn- [103] Khamzina L, Veilleux A, Bergeron S, Marette A. Increased
thesis associated with hypertrophy in cultured neonatal rat activation of the mammalian target of rapamycin pathway in
ventricular myocytes. FEBS Lett 1998;422:193 6. liver and skeletal muscle of obese rats: possible involvement
[88] Soesanto W, Lin HY, Hu E, Lefler S, Litwin SE, Sena S, et al. in obesity-linked insulin resistance. Endocrinology
Mammalian target of rapamycin is a critical regulator of car- 2005;146:1473 81.
diac hypertrophy in spontaneously hypertensive rats. [104] Drake JC, Alway SE, Hollander JM, Williamson DL. AICAR
Hypertension 2009;54:1321 7. treatment for 14 days normalizes obesity-induced dysregula-
[89] Gao XM, Wong G, Wang B, Kiriazis H, Moore XL, Su YD, et al. tion of TORC1 signaling and translational capacity in fasted
Inhibition of mTOR reduces chronic pressure-overload cardiac skeletal muscle. Am J Physiol Regul Integr Comp Physiol
hypertrophy and fibrosis. J Hypertens 2006;24:1663 70. 2010;299:R1546 54.
[90] Kemi OJ, Ceci M, Wisloff U, Grimaldi S, Gallo P, Smith GL, [105] Ranieri SC, Fusco S, Panieri E, Labate V, Mele M, Tesori V,
et al. Activation or inactivation of cardiac Akt/mTOR signal- et al. Mammalian life-span determinant p66shcA mediates
ing diverges physiological from pathological hypertrophy. obesity-induced insulin resistance. Proc Natl Acad Sci U S A
J Cell Physiol 2008;214:316 21. 2010;107:13420 5.
[91] Shioi T, McMullen JR, Tarnavski O, Converso K, Sherwood MC, [106] Um SH, Frigerio F, Watanabe M, Picard F, Joaquin M, Sticker M,
Manning WJ, et al. Rapamycin attenuates load-induced cardiac et al. Absence of S6K1 protects against age- and diet-induced
hypertrophy in mice. Circulation 2003;107:1664 70. obesity while enhancing insulin sensitivity. Nature
[92] Zhang D, Contu R, Latronico MV, Zhang J, Rizzi R, Catalucci 2004;431:200 5.
D, et al. MTORC1 regulates cardiac function and myocyte sur- [107] Sung MM, Koonen DP, Soltys CL, Jacobs RL, Febbraio M,
vival through 4E-BP1 inhibition in mice. J Clin Invest Dyck JR. Increased CD36 expression in middle-aged mice con-
2010;120:2805 16. tributes to obesity-related cardiac hypertrophy in the absence
[93] Tsukamoto O, Minamino T, Okada K, Shintani Y, Takashima S, of cardiac dysfunction. J Mol Med (Berl) 2011;89:459 69.
Kato H, et al. Depression of proteasome activities during the [108] Turdi S, Kandadi MR, Zhao J, Huff AF, Du M, Ren J.
progression of cardiac dysfunction in pressure-overloaded Deficiency in AMP-activated protein kinase exaggerates high
heart of mice. Biochem Biophys Res Commun fat diet-induced cardiac hypertrophy and contractile dysfunc-
2006;340:1125 33. tion. J Mol Cell Cardiol 2011;50:712 22.
[94] Harston RK, McKillop JC, Moschella PC, Van LA, Quinones LS, [109] Tremblay F, Brule S, Hee US, Li Y, Masuda K, Roden M, et al.
Baicu CF, et al. Rapamycin treatment augments both protein Identification of IRS-1 Ser-1101 as a target of S6K1 in nutrient-
ubiquitination and Akt activation in pressure-overloaded rat and obesity-induced insulin resistance. Proc Natl Acad Sci
myocardium. Am J Physiol Heart Circ Physiol 2011;300: USA 2007;104:14056 61.
H1696 706. [110] Duvel K, Yecies JL, Menon S, Raman P, Lipovsky AI, Souza AL,
[95] Vander HE, Lee SI, Bandhakavi S, Griffin TJ, Kim DH. Insulin et al. Activation of a metabolic gene regulatory network down-
signalling to mTOR mediated by the Akt/PKB substrate stream of mTOR complex 1. Mol Cell 2010;39:171 83.
PRAS40. Nat Cell Biol 2007;9:316 23. [111] Porstmann T, Santos CR, Griffiths B, Cully M, Wu M,
[96] Shen WH, Chen Z, Shi S, Chen H, Zhu W, Penner A, et al. Leevers S, et al. SREBP activity is regulated by mTORC1 and
Cardiac restricted overexpression of kinase-dead mammalian contributes to Akt-dependent cell growth. Cell Metab
target of rapamycin (mTOR) mutant impairs the mTOR- 2008;8:224 36.
mediated signaling and cardiac function. J Biol Chem [112] Cybulski N, Polak P, Auwerx J, Ruegg MA, Hall MN. mTOR
2008;283:13842 9. complex 2 in adipose tissue negatively controls whole-body
[97] Song X, Kusakari Y, Xiao CY, Kinsella SD, Rosenberg MA, growth. Proc Natl Acad Sci U S A 2009;106:9902 7.
Scherrer-Crosbie M, et al. mTOR attenuates the inflammatory [113] Polak P, Cybulski N, Feige JN, Auwerx J, Ruegg MA, Hall MN.
response in cardiomyocytes and prevents cardiac dysfunction Adipose-specific knockout of raptor results in lean mice with
in pathological hypertrophy. Am J Physiol Cell Physiol enhanced mitochondrial respiration. Cell Metab
2010;299:C1256 66. 2008;8:399 410.
[98] McMullen JR, Shioi T, Zhang L, Tarnavski O, Sherwood MC, [114] Brown NF, Stefanovic-Racic M, Sipula IJ, Perdomo G. The
Dorfman AL, et al. Deletion of ribosomal S6 kinases does not mammalian target of rapamycin regulates lipid metabolism in
attenuate pathological, physiological, or insulin-like growth primary cultures of rat hepatocytes. Metabolism
factor 1 receptor-phosphoinositide 3-kinase-induced cardiac 2007;56:1500 7.
hypertrophy. Mol Cell Biol 2004;24:6231 40. [115] Peng T, Golub TR, Sabatini DM. The immunosuppressant
[99] Shende P, Plaisance I, Morandi C, Pellieux C, Berthonneche C, rapamycin mimics a starvation-like signal distinct from amino
Zorzato F, et al. Cardiac raptor ablation impairs adaptive acid and glucose deprivation. Mol Cell Biol 2002;22:5575 84.

IV. mTOR IN CARDIOVASCULAR DEVELOPMENT AND DISEASE


REFERENCES 261
[116] Zhou G, Myers R, Li Y, Chen Y, Shen X, Fenyk-Melody J, et al. [133] Guo R, Zhang Y, Turdi S, Ren J. Adiponectin knockout accent-
Role of AMP-activated protein kinase in mechanism of metfor- uates high fat diet-induced obesity and cardiac dysfunction:
min action. J Clin Invest 2001;108:1167 74. role of autophagy. Biochim Biophys Acta 2013;1832:1136 48.
[117] Hagiwara A, Cornu M, Cybulski N, Polak P, Betz C, Trapani [134] Xu X, Hua Y, Nair S, Zhang Y, Ren J. Akt2 knockout preserves
F, et al. Hepatic mTORC2 activates glycolysis and lipogenesis cardiac function in high-fat diet-induced obesity by rescuing
through Akt, glucokinase, and SREBP1c. Cell Metab cardiac autophagosome maturation. J Mol Cell Biol
2012;15:725 38. 2013;5:61 3.
[118] Kim JE, Chen J. Regulation of peroxisome proliferator- [135] Xu X, Kobayashi S, Chen K, Timm D, Volden P, Huang Y,
activated receptor-gamma activity by mammalian target of et al. Diminished autophagy limits cardiac injury in mouse
rapamycin and amino acids in adipogenesis. Diabetes models of type 1 diabetes. J Biol Chem 2013;288:18077 92.
2004;53:2748 56. [136] Das A, Durrant D, Koka S, Salloum FN, Xi L, Kukreja RC.
[119] Zhang HH, Huang J, Duvel K, Boback B, Wu S, Squillace RM, Mammalian target of rapamycin (mTOR) inhibition with rapa-
et al. Insulin stimulates adipogenesis through the Akt-TSC2- mycin improves cardiac function in type 2 diabetic mice:
mTORC1 pathway. PLoS One 2009;4:e6189. potential role of attenuated oxidative stress and altered con-
[120] Cho HJ, Park J, Lee HW, Lee YS, Kim JB. Regulation of adipo- tractile protein expression. J Biol Chem 2014;289:4145 60.
cyte differentiation and insulin action with rapamycin. [137] Deepa SS, Walsh ME, Hamilton RT, Pulliam D, Shi Y, Hill S,
Biochem Biophys Res Commun 2004;321:942 8. et al. Rapamycin modulates markers of mitochondrial biogen-
[121] Gagnon A, Lau S, Sorisky A. Rapamycin-sensitive phase of esis and fatty acid oxidation in the adipose tissue of db/db
3T3-L1 preadipocyte differentiation after clonal expansion. J mice. J Biochem Pharmacol Res 2013;1:114 23.
Cell Physiol 2001;189:14 22. [138] Chambers KT, Leone TC, Sambandam N, Kovacs A, Wagg CS,
[122] Clavijo LC, Pinto TL, Kuchulakanti PK, Torguson R, Chu WW, Lopaschuk GD, et al. Chronic inhibition of pyruvate dehydro-
Satler LF, et al. Metabolic syndrome in patients with acute genase in heart triggers an adaptive metabolic response. J Biol
myocardial infarction is associated with increased infarct size Chem 2011;286:11155 62.
and in-hospital complications. Cardiovasc Revasc Med [139] Boudina S, Sena S, Theobald H, Sheng X, Wright JJ, Hu XX, et al.
2006;7:7 11. Mitochondrial energetics in the heart in obesity-related diabetes:
[123] Goldberg IJ, Trent CM, Schulze PC. Lipid metabolism and tox- direct evidence for increased uncoupled respiration and activa-
icity in the heart. Cell Metab 2012;15:805 12. tion of uncoupling proteins. Diabetes 2007;56:2457 66.
[124] Pulakat L, DeMarco VG, Ardhanari S, Chockalingam A, Gul [140] Boudina S, Abel ED. Diabetic cardiomyopathy, causes and
R, Whaley-Connell A, et al. Adaptive mechanisms to compen- effects. Rev Endocr Metab Disord 2010;11:31 9.
sate for overnutrition-induced cardiovascular abnormalities. [141] Cai L, Wang Y, Zhou G, Chen T, Song Y, Li X, et al.
Am J Physiol Regul Integr Comp Physiol 2011;301:R885 95. Attenuation by metallothionein of early cardiac cell death via
[125] Lee S, Zhang H, Chen J, Dellsperger KC, Hill MA, Zhang C. suppression of mitochondrial oxidative stress results in a pre-
Adiponectin abates diabetes-induced endothelial dysfunction vention of diabetic cardiomyopathy. J Am Coll Cardiol
by suppressing oxidative stress, adhesion molecules, and 2006;48:1688 97.
inflammation in type 2 diabetic mice. Am J Physiol Heart Circ [142] Matsushima S, Kinugawa S, Ide T, Matsusaka H, Inoue N,
Physiol 2012;303:H106 15. Ohta Y, et al. Overexpression of glutathione peroxidase
[126] Zhang H, Dellsperger KC, Zhang C. The link between meta- attenuates myocardial remodeling and preserves diastolic
bolic abnormalities and endothelial dysfunction in type 2 dia- function in diabetic heart. Am J Physiol Heart Circ Physiol
betes: an update. Basic Res Cardiol 2012;107:237. 2006;291:H2237 45.
[127] Zhang P, Hu X, Xu X, Fassett J, Zhu G, Viollet B, et al. AMP [143] Matsushima S, Ide T, Yamato M, Matsusaka H, Hattori F,
activated protein kinase-alpha2 deficiency exacerbates Ikeuchi M, et al. Overexpression of mitochondrial peroxiredoxin-
pressure-overload-induced left ventricular hypertrophy and 3 prevents left ventricular remodeling and failure after myocar-
dysfunction in mice. Hypertension 2008;52:918 24. dial infarction in mice. Circulation 2006;113:1779 86.
[128] Paiva MA, Rutter-Locher Z, Goncalves LM, Providencia LA, [144] Shen X, Zheng S, Metreveli NS, Epstein PN. Protection of car-
Davidson SM, Yellon DM, et al. Enhancing AMPK activation diac mitochondria by overexpression of MnSOD reduces dia-
during ischemia protects the diabetic heart against reperfusion betic cardiomyopathy. Diabetes 2006;55:798 805.
injury. Am J Physiol Heart Circ Physiol 2011;300:H2123 34. [145] Pham CG, Bubici C, Zazzeroni F, Papa S, Jones J, Alvarez K,
[129] Jessen N, Koh HJ, Folmes CD, Wagg C, Fujii N, Lofgren B, et al. Ferritin heavy chain upregulation by NF-kappaB inhi-
et al. Ablation of LKB1 in the heart leads to energy depriva- bits TNFalpha-induced apoptosis by suppressing reactive
tion and impaired cardiac function. Biochim Biophys Acta oxygen species. Cell 2004;119:529 42.
2010;1802:593 600. [146] Torti FM, Torti SV. Regulation of ferritin genes and protein.
[130] Wang CY, Kim HH, Hiroi Y, Sawada N, Salomone S, Blood 2002;99:3505 16.
Benjamin LE, et al. Obesity increases vascular senescence and [147] Omiya S, Hikoso S, Imanishi Y, Saito A, Yamaguchi O, Takeda
susceptibility to ischemic injury through chronic activation of T, et al. Downregulation of ferritin heavy chain increases labile
Akt and mTOR. Sci Signal 2009;2:ra11. iron pool, oxidative stress and cell death in cardiomyocytes.
[131] Li ZL, Woollard JR, Ebrahimi B, Crane JA, Jordan KL, Lerman J Mol Cell Cardiol 2009;46:59 66.
A, et al. Transition from obesity to metabolic syndrome is [148] Volkers M, Doroudgar S, Nguyen N, Konstandin MH, Quijada
associated with altered myocardial autophagy and apoptosis. P, Din S, et al. PRAS40 prevents development of diabetic car-
Arterioscler Thromb Vasc Biol 2012;32:1132 41. diomyopathy and improves hepatic insulin sensitivity in obe-
[132] Li ZL, Ebrahimi B, Zhang X, Eirin A, Woollard JR, Tang H, sity. EMBO Mol Med 2014;6:57 65.
et al. Obesity-metabolic derangement exacerbates cardio- [149] Alegria JR, Miller TD, Gibbons RJ, Yi QL, Yusuf S. Infarct size,
myocyte loss distal to moderate coronary artery stenosis in ejection fraction, and mortality in diabetic patients with acute
pigs without affecting global cardiac function. Am J Physiol myocardial infarction treated with thrombolytic therapy. Am
Heart Circ Physiol 2014;306:H1087 101. Heart J 2007;154:743 50.

IV. mTOR IN CARDIOVASCULAR DEVELOPMENT AND DISEASE


262 15. ROLE OF mTOR SIGNALING IN CARDIOPROTECTION

[150] Cubbon RM, Wheatcroft SB, Grant PJ, Gale CP, Barth JH, is not restored by insulin in the rat heart: phosphorylated signal
Sapsford RJ, et al. Temporal trends in mortality of patients transducer and activator of transcription 3- and phospha-
with diabetes mellitus suffering acute myocardial infarction: a tidylinositol 3-kinase-mediated inhibition. Anesthesiology
comparison of over 3000 patients between 1995 and 2003. Eur 2011;114:1364 72.
Heart J 2007;28:540 5. [153] Hilfiker-Kleiner D, Hilfiker A, Fuchs M, Kaminski K, Schaefer
[151] Das A, Salloum FN, Filippone SM, Durrant DE, Rokosh G, A, Schieffer B, et al. Signal transducer and activator of tran-
Bolli R, et al. Inhibition of mammalian target of rapamycin scription 3 is required for myocardial capillary growth, control
protects against reperfusion injury in diabetic heart through of interstitial matrix deposition, and heart protection from
STAT3 signaling. Basic Res Cardiol 2015;110:31. ischemic injury. Circ Res 2004;95:187 95.
[152] Drenger B, Ostrovsky IA, Barak M, Nechemia-Arbely Y, Ziv E,
Axelrod JH. Diabetes blockade of sevoflurane postconditioning

IV. mTOR IN CARDIOVASCULAR DEVELOPMENT AND DISEASE

You might also like