You are on page 1of 16

JOURNAL OF THE AMERICAN COLLEGE OF CARDIOLOGY VOL. 69, NO.

15, 2017

ª 2017 BY THE AMERICAN COLLEGE OF CARDIOLOGY FOUNDATION ISSN 0735-1097/$36.00

PUBLISHED BY ELSEVIER http://dx.doi.org/10.1016/j.jacc.2017.01.064

THE PRESENT AND FUTURE

STATE-OF-THE-ART REVIEW

The Aging Cardiovascular System


Understanding It at the Cellular and Clinical Levels

Francesco Paneni, MD, PHD,a,b Candela Diaz Cañestro, MSC,a Peter Libby, MD, PHD,c Thomas F. Lüscher, MD,a,b
Giovanni G. Camici, PHDa,b

ABSTRACT

Cardiovascular disease (CVD) presents a great burden for elderly patients, their caregivers, and health systems.
Structural and functional alterations of vessels accumulate throughout life, culminating in increased risk of devel-
oping CVD. The growing elderly population worldwide highlights the need to understand how aging promotes CVD in
order to develop new strategies to confront this challenge. This review provides examples of some major unresolved
clinical problems encountered in daily cardiovascular practice as we care for elderly patients. Next, the authors
summarize the current understanding of the mechanisms implicated in cardiovascular aging, and the potential for
targeting novel pathways implicated in endothelial dysfunction, mitochondrial oxidative stress, chromatin remod-
eling, and genomic instability. Lastly, the authors consider critical aspects of vascular repair, including autologous
transplantation of bone marrow-derived stem cells in elderly patients. (J Am Coll Cardiol 2017;69:1952–67)
© 2017 by the American College of Cardiology Foundation.

A ge dominates risk factors for cardiovascular


disease (CVD) (1,2). Indeed, the advent of
contemporary treatments for acute coronary
syndromes and stroke have helped to extend life
increasing dramatically, particularly in low- and
middle-income countries (e.g., Chile, China, Iran,
and Russia).
Although aging presents an array of disorders (5),
expectancy (3). Although an enormous success from CVD carries the greatest burden for the older popu-
an individual perspective, the resultant demographic lation, their care givers, and health systems (6). Cor-
shift presents one of the greatest challenges for the onary heart disease (CHD) associates strongly with
social and health care systems worldwide (4). The age, and is the leading cause of death in Europe and
population over 65 years of age will double from the United States (7–9). The prevalence of CVD in-
12% in 2010 to 22% in 2040 (2). Indeed, by 2020, the creases in people >65 years of age, especially in those
number of people 60 years of age and older will sur- >80 years of age, and will increase by w10% over the
pass the number of children below 5 years of age. next 20 years (2). From 2010 to 2030, an additional
The pace of population aging around the world is 27 million people will have hypertension, 8 million

From the aCenter for Molecular Cardiology, University of Zurich, Zurich, Switzerland; bUniversity Heart Center, Cardiology,
University Hospital Zurich, Zurich, Switzerland; and the cBrigham and Women’s Hospital, Division of Cardiovascular Medicine,
Boston, Massachusetts. The present work was supported by the Swiss National Science Foundation (Drs. Lüscher and Camici) and
the Alfred and Annemarie von Sick Grants for Translational and Clinical Research Cardiology and Oncology (Dr. Camici), an
Listen to this manuscript’s unrestricted research grant by Pfizer, Inc. (Dr. Lüscher), the Foundation for Cardiovascular Research–Zurich Heart House and the
audio summary by U.S. National Institutes of Health (RO1 HL080472), and from the RRM Charitable Fund (Dr. Libby). Drs. Paneni and Camici are
JACC Editor-in-Chief recipients of a Sheikh Khalifa’s Foundation Assistant Professorship at the Faculty of Medicine, University of Zurich. Dr. Libby has
Dr. Valentin Fuster. been an unpaid consultant for Amgen, AstraZeneca, Esperion Therapeutics, Ionis Pharmaceuticals, Kowa Pharmaceuticals, Merck
& Co., Novartis, Pfizer, Sanofi-Regeneron, Takeda Pharmaceuticals, and XBiotech; served as a scientific advisory board member
for Amgen, Athera Biotechnologies, Corvidia Therapeutics, DalCor Pharmaceuticals, Interleukin Genetics, Kowa Pharmaceuticals,
Medimmune, and Novartis; and has received lab funding from Novartis. All other authors have reported that they have no
relationships relevant to the contents of this paper to disclose.

Manuscript received July 12, 2016; revised manuscript received January 23, 2017, accepted January 24, 2017.
JACC VOL. 69, NO. 15, 2017 Paneni et al. 1953
APRIL 18, 2017:1952–67 The Aging Cardiovascular System

will have CHD, 4 million will have stroke, and 3 determinant of myocardial oxygen re- ABBREVIATIONS

million will have heart failure due to the rapid accu- quirements. Chronic exposure to increased AND ACRONYMS

mulation of elders (2). systolic pressure leads to LV hypertrophy,


BM = bone marrow
Increased CVD prevalence also interplays with causing a further rise in myocardial oxygen
CAC = circulating angiogenic
frailty, a condition of increased vulnerability to demand. Mediators, such as TGF-b , angio-
cell
stressors. A meta-analysis that included 54,250 tensin II, and the mineralocorticoid aldoste-
CHD = coronary heart disease
elderly patients associated CVD with an odds ratio rone, contribute mechanistically to
CI = confidence interval
(OR) of 2.7 to 4.1 for prevalent frailty and an OR of 1.5 hypertrophy and fibrosis in the pressure-
COX = cyclooxygenase
for incident frailty in those without frailty at baseline overloaded LV. Thus, the systolic hyperten-
CV = cardiovascular
(10). Current projections predict an increase in ex- sion and decreased diastolic pressure associ-
penditures for CHD and heart failure of w200% over ated with aging yield a “perfect storm” of CVD = cardiovascular disease

the next 20 years, with stroke expected to contribute decreased oxygen supply in the face of eNOS = endothelial nitric
oxide synthase
the largest relative increase in annual medical costs of augmented oxygen demand. As coronary
EPC = endothelial progenitor
238% (2). These considerations highlight the urgency atherosclerosis also advances with age, this
cell
of understanding why age contributes crucially to the additional limitation to oxygen supply often
HFpEF = heart failure with
development of CVD in order to enable us to cope compounds the increased oxygen demand preserved ejection fraction
with the aging of the population. This review pro- typically encountered in the aging CV system
LTL = leukocyte telomere
vides clinical and experimental evidence to support (Figure 1). In addition to altered large artery length
the established link between aging and CVD. function, chronic hypertension promotes LV = left ventricular
remodeling of the myocardial microvascula- MI = myocardial infarction
CLINICAL ASPECTS OF CVD IN ture. Thickening of the tunica media of
MMP = matrix
ELDERLY PATIENTS myocardial arterioles can further impede LV metalloproteinase
perfusion and impair vasomotion. These NF-kB = nuclear factor kappa-B
A number of age-related conditions present particular consequences of the increased systolic and OR = odds ratio
challenges to the clinical care of our cardiovascular decreased diastolic pressure that typically
PBMC = peripheral blood
(CV) patients. The accumulation of elderly in- accompany aging conspire to cause myocar- mononuclear cell
dividuals in our population underscores the growing dial ischemia. The regulation of arteriolar RAAS = renin-angiotensin-
importance of CV aging to practitioners. remodeling in the myocardium subjected to aldosterone system

SYSTOLIC HYPERTENSION AND WIDENED PULSE increased systolic and decreased diastolic ROS = reactive oxygen species

PRESSURE. With aging, the aorta stiffens due to pressure likely involves the same pathways RR = relative risk
increased collagen and decreased elastin (Figure 1). implicated in generation of aortic stiffness. TGF = transforming growth
Augmented transforming growth factor (TGF)-b We lack sufficient understanding of the pri- factor

activity favors the accumulation of collagen in the mary age-related triggers for these patho- TNF = tumor necrosis factor

aortic wall. The activity of various elastases, physiological processes that contribute VSMC = vascular smooth
including matrix metalloproteinases (MMPs), such as enormously to CV complications in the aging muscle cell

MMP-9 and MMP-12, as well as overexpression of the population. wtTTR = wild-type


transthyretin
cysteine proteinases cathepsins S, K, and L, and the Fortunately, some interventions can
serine proteinase neutrophil elastase, elaborated by improve outcomes in patients with isolated
inflammatory cells, can all contribute to depletion of systolic hypertension. The SHEP (Systolic
elastin (11). These alterations in the aorta’s extracel- Hypertension in the Elderly), Syst-Eur (Systolic Hy-
lular matrix contribute importantly to its loss of pertension in Europe), Syst-China (Systolic Hyper-
distensibility. This increased stiffness raises reflected tension in China), and other studies offer a rich
waves and elevates systolic pressure. Yet diastolic database affirming the ability of pharmacological
pressure tends to decline with age. As aortic pulse treatment of individuals in their 60s or 70s to reduce
wave velocity increases, pulse pressure rises (12). substantially stroke and total mortality, with lesser
Indeed, pulse pressure is an independent risk factor benefit for ischemic cardiac events (16). Avoiding
for CV events (13). Isolated systolic hypertension ac- excessive sodium intake may provide an additional,
counts for the majority of uncontrolled hypertension nonpharmacological intervention for control of hy-
in Americans over 50 years of age (14,15). pertension in older individuals (17,18). Some have
The fall in diastolic pressure decreases the drive for raised concerns regarding the safety of aggressive
coronary perfusion that occurs primarily during lowering of blood pressure in elderly patients,
diastole, favoring the development of myocardial particularly those with concomitant coronary artery
ischemia. The increased systolic pressure with age disease (19). Indeed, a J-shaped curve relating CV
increases left ventricular (LV) afterload, a major outcomes to blood pressure may pertain to this
1954 Paneni et al. JACC VOL. 69, NO. 15, 2017

The Aging Cardiovascular System APRIL 18, 2017:1952–67

F I G U R E 1 Pathophysiology of Aortic-LV Dynamics in the Aging CV System

Ang II ¼ angiotensin II; BP ¼ blood pressure; CV ¼ cardiovascular; HFpEF ¼ heart failure with preserved ejection fraction; HFrEF ¼ heart failure with reduced ejection
fraction; LV ¼ left ventricular; MMP ¼ matrix metalloproteinase; MVO2 ¼ myocardial oxygen consumption; TGF ¼ transforming growth factor.

population (20–22). Yet the HYVET (Hypertension in or attenuate the increased risk of CVD in elderly
the Very Elderly Trial) and SPRINT (Systolic Blood people.
Pressure Intervention Trial) studies confirmed the Several recent (pre-)clinical studies have estab-
relative safety and efficacy of antihypertensive lished key vascular modifications occurring with
treatment in the older population, although recent aging (31,32), identifying 2 main features: generalized
data suggest maintaining diastolic pressure above endothelial dysfunction and central arterial stiffness.
70 mm Hg in those with established coronary artery With regard to the former, vascular aging alters
disease (21–25). Clinicians should tailor the aggres- the function of the endothelium, the cells that line
siveness of antihypertensive treatment of geriatric the lumen of blood vessels. Endothelial dysfunction
patients on an individualized basis, depending on the includes reduced vasodilatory and antithrombotic
status of the coronary arteries, frailty, the integrity of properties, with an increase in oxidative stress
autonomic function, and other variables (26). and inflammatory cytokines (33–35) favoring athero-
In addition to reducing stroke, a major impediment genesis and thrombosis, and predisposing to CVD
to independent living and function in older patients, (36). Human and experimental studies concur
antihypertensive therapy may limit the development that diminished bioavailability of nitric oxide (NO), a
of dementing illnesses, as shown in the Syst-Eur key mediator of vasorelaxation and antiatherogenic
trial (27). Decreased dementia and cognitive decline processes, underlies age-dependent endothelial
accrue with longer duration of antihypertensive dysfunction (37,38). Reduced NO bioavailability can
treatment (28). An asymmetric loss to follow-up of occur due to decreased synthesis or increased
individuals with impaired cognition may have biased degradation of NO. Under normal conditions, endo-
the results of dementia in the SHEP study to the thelial nitric oxide synthase (eNOS) produces NO from
null (29). L -arginine in the presence of the cofactor tetrahy-
drobiopterin (BH4) (39). Although studies differ
VASCULAR AGING: DEFINITION, PATHOPHYSIOLOGY, regarding eNOS protein expression with age (34,40,41),
AND IMPACT. Alterations in the structure and function recent data suggest an age-related alteration in
of arteries accompany aging, and contribute to eNOS function, referred to as eNOS uncoupling (42).
increased risks of developing CVD (9,30). Accord- This effect derives, at least in part, from a decrease in
ingly, understanding the mechanisms by which age BH4 availability, resulting in impaired NO release and
affects the vasculature should position us to prevent increased production of the highly pro-oxidant
JACC VOL. 69, NO. 15, 2017 Paneni et al. 1955
APRIL 18, 2017:1952–67 The Aging Cardiovascular System

superoxide anion (O 2) (33). Furthermore, the avail- function that disrupts arterial homeostasis, favoring
ability of the eNOS substrate, L -arginine, may also chronic activation of pro-oxidant and proin-
restrict NO production. In this regard, the activity of flammatory pathways, and a shift toward a vasocon-
arginase, an enzyme that competes with eNOS for strictor profile, predisposing to CVD and adverse
L -arginine, increases with age, providing a plausible events (51).
mechanism for the decreased synthesis of NO (43). Increased stiffness of central arteries, particularly
Nevertheless, most of this mechanistic evidence has of the thoracic aorta, also characterizes human
emerged from animal studies, highlighting the need vascular aging (52) (Figure 1). Human and animal
for more human research regarding age-dependent studies suggest that the reduced distensibility of
limitation of NO synthesis. large elastic arteries results primarily from the loss of
Additionally, aging may increase NO degradation elastic fibers, as well as an increase in collagen (53).
(34) due to elevated concentrations of reactive oxy- Mechanistically, the age-related increase in elastin
gen species (ROS), mediated, in part, by chronic degradation may result from augmented activity of
inflammation, constituting a vicious cycle that de- proteases with elastinolytic activity, including certain
pletes NO (35). For example, age-dependent increases MMPs and cysteinyl cathepsins, enzymes that, in
in vascular and blood-borne tumor necrosis factor turn, are regulated by inflammatory mediators
(TNF)-a , associates with increased expression of (54,55). Collagen catabolism falls in aging arteries.
nicotinamide adenine dinucleotide phosphate The accumulation of collagen modified by advanced
(NADPH) oxidase (Figure 2). Augmented NADPH oxi- glycation end-products (AGEs) (condensates of
dase activity yields overproduction of O 2, which, in glucose produced by nonenzymatic reactions that
turn, reacts with NO to form peroxynitrite (ONOO ), a form cross-links between collagen molecules) may
powerful oxidant involved in nitrosylation of anti- contribute to this age-related increase in arterial
oxidant enzymes and eNOS (34). Besides inflamma- collagen. AGE-mediated cross-links can confer resis-
tory cytokines, the renin-angiotensin-aldosterone tance to enzymatic degradation, and thus interfere
system (RAAS) may contribute to the age-associated with collagenolysis (56). In addition, increased ac-
increase in NO inactivation. With aging, RAAS activ- tivity of TGF- b with aging stimulates the synthesis of
ity and the concentration of angiotensin II, a principal interstitial collagen by vascular smooth muscle cells
RAAS effector, rise, increasing the production of ROS (VSMCs), and thereby augments arterial stiffness (57).
by activating NADPH oxidase (44). In turn, age- Likewise, increased activity of the RAAS may
related increased ROS production may promote augment collagen synthesis and heighten elastolysis
vascular inflammation (45). Indeed, hydrogen (58).
peroxide (H2O 2) activates nuclear factor kappa-B Endothelial dysfunction and arterial stiffness are
(NF- k B), which augments the transcription of proin- closely interconnected mediators of age-dependent
flammatory genes, leading to increased expression of vascular dysfunction (59). The stiffer the artery, the
TNF- a , interleukin 6, chemokines, and adhesion greater the exposure of the endothelium to hemody-
molecules implicated in atherogenesis (Figure 2). namic load, promoting endothelial activation,
Increased vasoconstrictor tone may compound age- inflammation, and damage (60). Age-related re-
dependent impairment in endothelial-dependent ductions in NO bioavailability described earlier may
vasodilatation. Human aging associates with an in- contribute to impaired arterial distensibility via
crease in circulating and arterial wall levels of changes in smooth muscle tone (61). Novel therapies
endothelin(ET)-1, a powerful vasoconstrictor impli- aimed at attenuating age-related CVD could target
cated in impaired endothelium-dependent dilation these features of vascular aging, with particular
(40,46). Moreover, evidence from rodent endothelial emphasis on endothelial dysfunction and changes in
cells indicates that concentrations of cyclooxygenase the arterial extracellular matrix that cause central
(COX)-derived eicosanoids also change considerably arterial stiffness.
with age. With age, the COX-derived eicosanoids
associated with vasoconstriction and thrombosis rise HEART FAILURE WITH PRESERVED AND REDUCED
(i.e., prostaglandin H 2 [PGH2], thromboxane A 2 EJECTION FRACTION. Related etiologically to both
[TxA2]. and prostaglandin F2a [PGF 2a ]), whereas the acquisition of aortic stiffness and remodeling of
prostaglandin I 2 (PGI 2, prostacyclin) falls (47,48). the myocardium, its extracellular matrix, and its
Furthermore, impaired endothelial-dependent res- microvasculature, heart failure with preserved ejec-
ponsiveness to prostaglandins accompanies aging in tion fraction (HFpEF) plagues elderly patients,
both humans and animals (49,50). Thus, aging asso- particularly women. The risk of developing HFpEF
ciates with a concerted impairment in endothelial skyrockets with age (62,63). Indeed, hypertrophy
1956 Paneni et al. JACC VOL. 69, NO. 15, 2017

The Aging Cardiovascular System APRIL 18, 2017:1952–67

F I G U R E 2 Schematic Representation of Molecular Pathways Involved in Arterial Aging

Aged endothelial cells down-regulate JunD and SIRT1expression, leading to pro-oxidant and proinflammatory gene expression. Increased ROS and inflammatory
cytokines reduce NO availability. In parallel, age-related up-regulation of angiotensin II and COX-derived eicosanoids results in augmented vasoconstrictor
mediators, such as ET-1, TxA2, and PGF2a. This imbalance between dilator and constrictor factors prompts impaired vascular function. Furthermore, increased
MMP expression in VSMCs induces changes in structural components of the arterial wall (e.g., decreased elastin/collagen ratio), contributing to arterial
stiffness. AA ¼ arachidonic acid; ALDH-2 ¼ aldehyde dehydrogenase; AMPK ¼ 50 adenosine monophosphate-activated protein kinase; Arg II ¼ arginase II;
AT-1 ¼ angiotensin II receptor type 1; BH4 ¼ tetrahydrobiopterin; Ca2þ ¼ calcium ions; cGMP ¼ cyclic guanosine monophosphate; COX ¼ cyclooxygense; DP ¼
prostaglandin D2 receptor; ecSOD ¼ extracellular superoxide dismutase; EPI-4 ¼ prostaglandin E2 receptor 1-4; ETA ¼ endothelin receptor A; FP ¼ prosta-
glandin F receptor; GC ¼ guanylate cyclase; GTP ¼ guanosine triphosphate; ICAM ¼ intercellular adhesion molecule; IP ¼ prostacyclin receptor; MCP ¼
monocyte chemotactic protein; MnSOD ¼ manganese superoxide dismutase; NF-kB ¼ nuclear factor kB; NO ¼ nitric oxide; PGD2 ¼ prostaglandin D2; PGF2a ¼
prostaglandin F 2a; PGI2 ¼ prostacyclin; PKG ¼ protein kinase G; ROS ¼ reactive oxygen species; TNF- a ¼ tumor necrosis factor alpha; TP ¼ thromboxane
receptor; TxA2 ¼ thromboxane A2; VCAM ¼ vascular cell adhesion protein; VSMC ¼ vascular smooth muscle cell; other abbreviations as in Figure 1.
JACC VOL. 69, NO. 15, 2017 Paneni et al. 1957
APRIL 18, 2017:1952–67 The Aging Cardiovascular System

and fibrosis of the aged LV can impair lusitropy. people over 80 years of age have wtTTR amyloid de-
Practitioners generally acknowledge the increasing posits in their hearts at autopsy. Although the clinical
prevalence of HFpEF, its accentuation in elderly pa- significance of this finding remains uncertain, it is
tients, and its adverse outcomes in our aging patient highly likely that with the aging of the population and
population, with regard, not only to morbidity, but the advent of novel diagnostic modalities, wtTTR
also to reduced quality of life and increased resource amyloidosis will become better recognized as a
utilization (64). Unfortunately, we possess few (if contributor to heart failure in the older population.
any) evidence-based interventions to stem the Amyloidosis due to wtTTR requires differentiation
development or consequences of HFpEF. Studies in from amyloid light-chain amyloidosis, because the
progress of neurohumoral blockade may help to fill prognosis and treatment diverge substantially.
this important gap in our therapeutic approach to Although we currently lack effective therapy for
HFpEF in our elderly patients (65). However, treating amyloidosis, promising imaging technologies
myocardial ischemia, due to either myocardial should provide evolving tools to evaluate novel
infarction (MI) (ischemic cardiomyopathy) or sub- therapies currently in development or under inves-
endocardial ischemia in the hypertrophied LV, can tigation (72).
predispose to heart failure with reduced ejection FRAILTY AND SARCOPENIA IN THE AGING
fraction. Thus, aging can promote the development of POPULATION. Frailty and loss of muscle mass and
both major forms of heart failure that constitute a key function (sarcopenia) present a major challenge to
challenge to quality of life of aging individuals, as maintenance of independence, mobility, quality of
well as an enormous drain on health care resources life, and the ability of our CV patients to undergo
(Figure 1). surgery and other interventions (such as trans-
VALVULAR AND CARDIAC SKELETON CALCIFICATION. With catheter valve procedures) successfully. Moreover,
age, calcium in the axial skeleton particularly tends to the ability to tolerate CV therapies may decline with
decline, while accumulating in CV structures. frailty and sarcopenia, as discussed earlier for phar-
Appreciation has increased that conditions such as macological treatment of hypertension. We lack def-
calcific aortic stenosis actually reflect a systemic inite criteria for defining sarcopenia and frailty
process (66). Evidence points to inflammation as a (73,74), which associate with both osteoporosis and
potential physiological contributor to CV calcifica- obesity (75). Potential pathophysiological pathways
tion. Strong human genetic data also implicate lip- that link these apparently disparate conditions
oprotein(a) in the pathogenesis of aortic valve include proinflammatory cytokines. Further clarity in
calcification (67). Despite initial enthusiasm, clinical diagnosis and pathophysiological mechanisms would
trials have not substantiated that statin therapy can be highly desirable, given the increase in the elderly
limit progression of aortic valvular calcification. population affected by CV disease, and the limita-
Indeed, statins tend to accelerate coronary arterial tions these conditions present to patients and their
calcification (68,69). Given the pressing clinical management. Increased physical activity could pro-
importance of calcific aortic stenosis in geriatric pa- vide a nonpharmacological preventive approach to
tients, we urgently need a greater understanding of augmenting patient quality of life and function, and
its pathophysiology and preventive measures to could conceivably improve the ability to tolerate
mitigate its genesis, as once CV structures have pharmacological and interventional treatment of CV
calcified, therapeutic reversal of this process may conditions, hence improving CV outcomes.
prove elusive.
MOLECULAR FEATURES OF
AMYLOIDOSIS AND THE AGING CV SYSTEM. Certain AGE-RELATED CVD
forms of amyloidosis also particularly affect the older
population (70). The incidence of amyloid light-chain TELOMERES AND CELLULAR SENESCENCE. Accumulation
amyloidosis rises in older patients, related to the of senescent cells within the vascular wall and
increase in multiple myeloma with age. Cardiac heart can contribute to structural and functional
amyloidosis associated with wild-type transthyretin decline of the CV system with age (58). Considerable
(wtTTR) particularly affects the older population, evidence implicates telomere shortening in cellular
especially men. Although many have regarded car- senescence. Telomeres consist of repetitive nucleotide
diac amyloidosis as a rare condition, current data sequences (TTAGGG) at the ends of mammalian chro-
using novel imaging modalities show that some 13% mosomes, that preserve chromosome stability and
of cases of HFpEF in individuals $60 years of age integrity by preventing deterioration or fusion with
associate with wtTTR amyloidosis (71). At least 20% of neighboring chromosomes (76) (Central Illustration).
1958 Paneni et al. JACC VOL. 69, NO. 15, 2017

The Aging Cardiovascular System APRIL 18, 2017:1952–67

C E NT R AL IL L U STR AT IO N Molecular Hallmarks of CV Aging (Cellular Senescence, Genomic Instability,


Chromatin Remodeling, and Mitochondrial Oxidative Stress)

Paneni, F. et al. J Am Coll Cardiol. 2017;69(15):1952–67.

These molecular events are strongly interconnected and contribute to vascular and cardiac dysfunction in elderly patients. A thorough understanding of these complex
processes may offer pharmaceutical targets to improve human health during aging. CV ¼ cardiovascular; CVD ¼ cardiovascular disease; eNOS ¼ endothelial nitric
oxide synthase; NADPH ¼ nicotinamide adenine dinucleotide phosphate; SIRT1 ¼ sirtuin 1; VSMC ¼ vascular smooth muscle cell.

Each cell division shortens telomeric DNA until, at a setting of CVD. Leukocyte telomere length (LTL)
critical length, the cells lose capping function at the associates significantly with vascular cell senescence,
chromosomal ends, activating DNA damage check- aortic valve stenosis, CV risk factors (i.e., hyperten-
points, cell senescence, and eventually apoptosis. sion, type 2 diabetes, obesity, and smoking), and risk of
Telomere shortening has particular relevance in the atherothrombotic events. However, the causality of
JACC VOL. 69, NO. 15, 2017 Paneni et al. 1959
APRIL 18, 2017:1952–67 The Aging Cardiovascular System

these associations remains uncertain (77). Patients sarcoplasmic reticulum calcium, as well as calcium
with clinical and subclinical features of atherosclerosis release-induced I Ca calcium channel inactivation (84).
display reduced LTL compared with healthy These changes dampen mechanical efficiency and
controls, even after adjustment for relevant con- electrophysiological properties, and increase the risk
founders, such as age, sex, and race (78). In a recent of arrhythmias (i.e., atrial fibrillation) in the older
case-control study, individuals with shorter LTL population.
had a higher presence of ischemic (OR: 1.37; 95% con-
fidence interval [CI]: 1.06 to 1.77) and hemorrhagic MITOCHONDRIAL OXIDATIVE STRESS. Mitochondrial
stroke (OR: 1.48; 95% CI: 1.08 to 2.02) as compared with overproduction of ROS likely contributes to cellular
the highest tertile of telomere length (79). Moreover, senescence (9). This process ultimately leads to for-
patients with reduced LTL had a significantly mation of the highly reactive products O 2 or H 2O 2,
increased risk for both incident plaque (hazard ratio: whose accumulation and diffusion fosters senes-
1.49; 95% CI: 1.09 to 2.03) and plaque progression cence, DNA mutations, inflammation, and multiple
(hazard ratio: 1.61; 95% CI: 1.26 to 2.07) (80). A recent cell death pathways (Central Illustration) (82).
meta-analysis, including prospective and retrospec- The mitochondrial adaptor p66Shc gained
tive studies on the association between LTL and CHD increasing attention due to its pivotal role in ROS
(43,725 participants of whom 8,400 had CVD), generation and cellular apoptosis (Central Illustration)
revealed that patients with the shortest LTL had a (83). Cells lacking the p66 Shc gene have reduced
higher relative risk (RR) for CHD (RR: 1.54; 95% CI: 1.30 intracellular free radicals, and mice lacking p66Shc
to 1.83) and cerebrovascular disease (RR: 1.42; exposed to high oxidative stress have diminished
95% CI: 1.11 to 1.81) (81). systemic and intracellular ROS (83). Additionally,
Critical aspects associated with cellular senescence genetic deletion of p66Shc in the mouse extended
include age-dependent defects of adrenergic lifespan by 30% (84). Age-dependent alteration of
signaling and calcium handling. Plasma levels of p66Shc signaling profoundly affects CV homeostasis.
norepinephrine significantly increase with age, as the Indeed, deletion of p66 Shc in mice protects from sys-
result of reduced plasma clearance and increased temic and cerebral age-dependent endothelial
spillover from the tissues (81). A reduction of the dysfunction by virtue of decreased ROS production
catecholamine reuptake transporter localized in and conserved NO bioavailability (83,85,86). Our own
sympathetic nerve terminals also contributes to recent work also underscored the relevance of this
elevated catecholamine concentrations with aging. gene in the pathogenesis of stroke. In fact,
Together, these alterations progressively impair p66Shc -deficient mice display decreased ROS produc-
adrenergic responsiveness, resulting in b -adrenergic tion in the brain, and have reduced stroke size
desensitization. This phenomenon ultimately re- following ischemia-reperfusion brain injury (87). We
duces the number, affinity, and internalization of further demonstrated that even post-ischemic in vivo
b-adrenergic receptors (namely the b 1-adrenergic re- silencing of p66Shc prevents ischemia-reperfusion
ceptor subtype), coupled with abnormalities in brain injury in mice, mainly through preservation of
membrane adenylate cyclase activity or cellular pro- blood-brain barrier integrity (88). The observation
duction of cyclic adenosine monophosphate (82). that p66 Shc expression increases significantly in
Such defects of autonomic modulation favor chrono- stroke patients and correlates with neurological def-
tropic incompetence and reduced inotropic reserve of icits supports the clinical relevance of these experi-
the LV, thus affecting exercise tolerance. mental findings (88). Expression of p66 Shc also
Reduction of calcium reuptake by the myocardial increases in peripheral blood mononuclear cells
sarcoplasmic reticulum calcium adenosine triphos- (PBMCs) of patients with acute coronary syndrome
phatase (SERCA2a) is another key feature of car- and type 2 diabetes (89,90). The activation of this
diomyocyte aging, yielding impaired early diastolic protein by several CV risk factors, such as hypergly-
LV filling and a compensatory increase in atrial cemia, oxidized low-density lipoprotein, smoking,
contraction (83). Calcium transient amplitude de- and hypertension, further supports its contribution to
creases gradually with age, being 3.2-fold smaller in clinical CVD (9,83). Taken together, experimental and
myocytes from patients $75 years of age than in those clinical data strongly indicate p66 Shc as a potential
younger than 55 years of age (84). An age-related therapeutic target in the setting of age-related CVD
delay in propagation of the calcium transient from (Figure 2).
the sarcolemma to the cell center may also occur (84). The Activated Protein-1 (AP-1) transcription factor
Moreover, aged myocytes have reduced SERCA2 JunD has emerged as a mediator of oxidative stress in
expression, limiting the amount of releasable aging (Figure 2). Dimeric complexes from 3 main
1960 Paneni et al. JACC VOL. 69, NO. 15, 2017

The Aging Cardiovascular System APRIL 18, 2017:1952–67

families of DNA-binding proteins (Jun, Fos, and regulator of p66 Shc transcription, controlling acetyla-
ATF/CREB) assemble to form AP-1 transcription fac- tion of histone H3 bound to the p66Shc promoter (102).
tors with activities that depend critically on their Impaired SIRT1 activity also favors acetylation of
specific components and the cellular microenviron- NF- kB p65, leading to its nuclear translocation and
ment (91). JunD regulates cell growth and survival, transcription of inflammatory genes, as described
and protects cells against oxidative stress by modu- earlier (103). Moreover, SIRT1 represses detrimental
lating genes involved in antioxidant defense and ROS pathways of arterial aging, such as Forkhead box O
production (92). We recently reported that JunD falls (FOXO) 1, 3, and 4, thus preventing DNA damage, cell
during aging both in the mouse aorta and in PBMCs cycle arrest, and oxidative stress (104). SIRT1 deace-
from old as compared with young, healthy humans tylates LKB1, leading to activation of the final effector
(93). Young mice lacking JunD display premature enzyme 5 0 adenosine monophosphate-activated pro-
endothelial dysfunction and vascular senescence tein kinase, a central energy regulator involved in
similar to aged wild-type mice (93). Aortas from glucose homeostasis, maintenance of cellular adeno-
JunD / mice have augmentation of the aging sine triphosphate levels, and endothelial integrity
markers p53 and p16INK4a, reduced telomerase activ- via regulation of eNOS activity and autophagy
ity, and mitochondrial DNA damage. By contrast, (Figure 2) (105).
in vivo overexpression of JunD rescues vascular aging Klotho has also emerged as an important antiaging
features in old mice (93). Mechanistic experiments gene (106). Klotho protein functions as a circulating
revealed that age-associated reductions in JunD hormone that binds to a cell-surface receptor, and
cause an imbalance between oxidant (NADPH oxi- represses intracellular signals of insulin and insulin-
dase) and scavenger enzymes (namely, manganese like growth factor 1, key molecules preserving
superoxide dismutase and aldehyde dehydrogenase longevity (107). Genetic deletion of Klotho in mice
2), leading to early redox changes, mitochondrial associates with premature aging features, including
dysfunction, and vascular senescence (93). In accord vascular calcification, altered calcium/phosphate
with our findings, lack of JunD promotes pressure metabolism, and reduced lifespan (108). By contrast,
overload-induced apoptosis of myocytes, hypertro- Klotho overexpression prolongs lifespan in mice and
phic growth, and angiogenesis in the heart (94). protects against age-related CV and renal impairment
Ongoing screening of chemical libraries aim to iden- (109). Of clinical relevance, higher plasma Klotho
tify compounds capable of restoring JunD activity in levels associate independently with a lower likeli-
the vasculature and the heart. hood of CVD in humans, whereas low serum Klotho
The family of nicotinamide adenine dinucleotide- concentrations independently predict coronary artery
dependent proteins termed sirtuins have an estab- disease and arterial stiffness (106). Validation of
lished role in human aging (Figure 2) (9). A recent study Klotho as a clinically useful biomarker and a thera-
found that endogenous SIRT1 expression in VSMCs peutic target to counteract age-related CVD requires
correlated inversely with donor age (95). Age-related further study.
loss of SIRT1 correlated with functional deficits,
diminished stress response, reduced capacity for GENOMIC INSTABILITY. Accumulation of genetic
migration/proliferation, and increased senescence damage through the course of life likely contributes
(95). Moreover, activation of SIRT1 may promote importantly to aging (Central Illustration). Genome
preservation of endothelial cell function during aging. alterations fall into 3 main categories: 1) chemical
Hypercholesterolemic mice with endothelial-specific damage to genomic DNA; 2) mutations (e.g., addition,
SIRT1 overexpression or those exposed chronically to deletion, or substitution of bits of genetic code); and
a SIRT1 activator exhibit attenuated atherogenesis, 3) epigenetic alterations, which modify gene activity
whereas reduced SIRT1 activity results in greater foam without affecting DNA sequence. To cope with ge-
cell formation and atherosclerosis (96–98). The netic lesions, organisms have evolved highly profi-
observation that immunosuppressant drugs (i.e., cient DNA repair systems that can, in most cases,
sirolimus and everolimus) cause endothelial senes- restore the correct base pair sequence (110). Defects in
cence via SIRT1 inhibition supports these results (99). DNA repair nonetheless occur, and can contribute to
SIRT1 blockade also impairs eNOS functionality, cellular senescence and organ dysfunction (111).
whereas its activation improves endothelial NO avail- Genomic instability particularly affects the CV system
ability (100). Moreover, microRNA-217, an endogenous (112). Hutchinson–Gilford progeria syndrome, char-
SIRT1 inhibitor, triggers endothelial senescence by acterized by massive nuclear DNA damage, associates
suppressing SIRT1-dependent eNOS functionality with premature atherosclerosis and CVD, which lead
(101). Recent work identified SIRT1 as a master to fatal MI or stroke by an average age of 13 years
JACC VOL. 69, NO. 15, 2017 Paneni et al. 1961
APRIL 18, 2017:1952–67 The Aging Cardiovascular System

(113). Similarly, mice with genomic instability result- epigenetic modifications caused by environmental
ing from defective nucleotide excision repair genes stimuli can be inherited, thus contributing to early
ERCC1 and XPD recapitulate aging features, such as senescent traits and CVD in young adults (121).
endothelial cell senescence, vascular stiffness, and Chromatin modifications include DNA methylation
hypertension, at a very young age (114). CV aging in and post-translational histone modifications (Central
this setting results mostly from eNOS and sirtuin Illustration). DNA methylation, which involves the
deregulation, as well as augmented NADPH oxidase addition of a methyl group to DNA nucleotides, re-
(114). presses gene transcription by affecting chromatin
Beyond genetic diseases, a growing body of evi- accessibility to the transcriptional machinery. DNA
dence indicates that sporadic genomic mutations methylation progressively decreases with age in mice
accumulated across the lifetime represent a major and humans, whereas the rate of demethylation as-
underpinning of CVD (115). Several studies have shown sociates inversely with age (122,123). Large-scale
the presence of DNA damage in both circulating cells of analysis revealed unmethylated or partially methyl-
patients with atherosclerosis and in the plaques ated CpG islands in atherosclerotic plaques and leu-
themselves (116). PBMCs from patients with CHD have kocytes from both patients and atherosclerosis-prone
chromosomal damage and mitochondrial DNA de- mice (124). Changes in DNA methylation localize at
letions that correlate with disease severity (117). the promoters of several genes, including NOS, es-
Similarly, data from the AortaGen Consortium showed trogen receptors, collagen type XV alpha 1 (COL15A1),
that genetic variation in the nucleotide excision repair and the vascular endothelial growth factor receptor
components (namely, a single-nucleotide poly- (VEGFR). Hence, aberrant DNA methylation during
morphism [rs2029298] in the promoter region of the the lifetime results in altered transcription of critical
DNA damage-binding protein 2 [DDB2] gene) associ- regulatory genes for the induction of proatherogenic
ated strongly with carotid-femoral pulse wave velocity and senescent cellular functions (125).
(114). Genomic instability in senescent VSMCs in- Together with DNA-related changes, post-
creases phosphodiesterase type 1 (PDE1) expression, translational histone modifications, among them
with subsequent impairment of NO/cyclic guanosine methylation, acetylation, ubiquitination, and phos-
monophosphate signaling and endothelial dysfunc- phorylation, may cluster in different patterns to
tion. Human genetic studies consistently reveal sig- regulate chromatin architecture. Histone methylation
nificant associations of PDE1A single-nucleotide may result in different chromatin states, depending
polymorphisms with diastolic blood pressure and ca- on the methylated residue and the number of methyl
rotid intima-media thickness (118). Overall, extensive groups added (120). Methylation marks on histone
evidence shows that genomic damage accompanies CV H3 regulate lifespan, as well as vascular homeostasis
aging. With age, humans can accumulate somatic (126,127). For example, histone methylation by the
mutations that give rise to hematopoietic clones mammalian methyltransferase Set7 regulates endo-
associated not only with myelodysplastic syndromes thelial NF- kB signaling, a pivotal modulator of
and hematologic malignancies, but surprisingly, even inflammation and longevity (128,129). In addition,
more strongly with CV risk (119). Future research in methylation by Set7 fundamentally regulates key
this area should unveil new interventions aimed at lifespan determinants heavily involved in the modu-
preserving genome stability to alleviate age-related lation of CV homeostasis, namely SIRT1, FoxO3, and
CVD. p53 (130). Set7 expression increases in PBMCs of pa-
tients with type 2 diabetes, and significantly corre-
CHROMATIN MODIFICATIONS. Although many lates with NF-k B–mediated inflammation, oxidative
studies have focused on the genes that influence ag- stress, and endothelial dysfunction, supporting the
ing, nongenomic regulation of aging has gained clinical relevance of these observations (131). Histone
increasing attention. Growing evidence suggests that deacetylation by SIRT1 may also influence age-related
epigenetic modifications can derail transcriptional CV disease. Transgenic overexpression of SIRT1 im-
programs implicated in oxidative stress, inflamma- proves metabolic efficiency and endothelial function
tion, angiogenesis, and cellular metabolism, thus in aged mice (103). SIRT6 can prevent endothelial
fostering maladaptive pathways and features of dysfunction and atherosclerosis by epigenetic mod-
vascular aging (Central Illustration) (120). Epigenetic ulation of multiple atherosclerosis-related genes,
modifications acquired during life appear durable and including the proatherogenic gene, tumor necrosis
relatively stable, thus providing a molecular frame- factor superfamily member 4 (TNFSF4) (132). These
work through which the environment interacts with results shed light on how chromatin modifications
the genome to alter gene expression. Indeed, may regulate aging-related features, and identify
1962 Paneni et al. JACC VOL. 69, NO. 15, 2017

The Aging Cardiovascular System APRIL 18, 2017:1952–67

reversal of epigenetic modifications as an attractive studies have documented a reduced number of BM-
therapeutic target. derived cells and CACs in aged patients, as well as
in subjects exposed to modifiable CV risk factors
AGE-ASSOCIATED DEFECTS IN (139). Features of senescence, such as telomere
VASCULAR REPAIR shortening, genomic instability, and subsequent cell
cycle arrest, accompany this reduced cell number
IMPAIRMENT OF ANGIOGENESIS. Elderly individuals (140) (Figure 3). Kushner et al. (141) showed an
not only have increased incidence of stroke, periph- approximately 60% decline in telomerase activity in
eral artery disease, and MI, but also display worsened CACs of older men (56 to 67 years of age). Over-
outcomes than younger patients. Aged patients with expression of human telomerase reverse transcrip-
acute limb ischemia have higher mortality and an tase in CACs preserved telomerase activity, delayed
increased rate of limb amputation (133). Moreover, cell senescence, and improved age-related CAC
about 35% to 40% of elderly patients experience dysfunction in mice with ischemic hind limbs (142).
inadequate myocardial reperfusion post-MI, eventu- Increased production of ROS and decreased expres-
ally leading to adverse LV remodeling, heart failure, sion of antioxidant enzymes also produces senescent
and CV death (134). Available evidence strongly cell changes. Early endothelial progenitor cells (EPCs)
indicates an association of aging with several defects from elderly subjects show reduced activity of the
in angiogenesis. Elderly men display reduced capil- antioxidant enzyme glutathione peroxidase-1 in
lary density, which is associated with microvascular association with enhanced cell apoptosis (143).
disease, defective eNOS functionality, and impaired Increased angiotensin II may also stimulate ROS
insulin sensitivity (135). Furthermore, senescent production and cellular senescence in early EPCs
endothelial cells have lower proliferative capacity, (144). The mobilization of progenitor cells from the
decreased telomerase activity, and reduced produc- BM, and their homing and engraftment depend upon
tion of angiogenic growth factors, such as VEGF-A. a number of angiogenic factors, SDF1/CXCR4 inter-
Endothelial migration is also impaired, leading to action, and VEGF (145). We recently showed profound
reduced tube formation (133). Reduced hypoxia- dysregulation of the aging and longevity genes p66Shc
inducible factor 1 a (HIF1 a ) activity, mainly due to and JunD in old (as compared with young) EPCs,
increased degradation and reduced nuclear trans- promoting mitochondrial oxidative stress and
location by importin a , significantly contributes to impairing SDF-1 secretion (146). This study indicates
age-related impairment of angiogenesis after that altered gene expression trajectories during life
ischemia. Interestingly, adenoviral delivery of a strongly affect stem and progenitor cell functions.
constitutively active form of HIF1 a improved neo-
ROLE OF EPIGENETICS. Time-dependent accumula-
vascularization and rest pain in patients with critical
tion of adverse epigenetic modifications has emerged
limb ischemia, whereas it failed to improve walking
as a potential underpinning of stem cell dysfunction
performance in patients with intermittent claudica-
in aging (Figure 3). Transfer of epigenetic information
tion (136,137). The activity of PGC-1a , an emerging
(i.e., noncoding RNAs [ncRNAs]), which may occur
transcriptional coactivator that plays an essential role
either by direct cell-to-cell contact or from the
in hypoxia-driven angiogenesis, also declines with
bloodstream into the cell, may strongly influence
age (133). Dysregulation of angiogenic pathways is
phenotype and cell fate decisions (139). More recent
associated with an age-dependent reduction in the
studies showed that extracellular vesicles, including
number and functionality of stem and progenitor
microparticles, exosomes, and apoptotic bodies, can
cells (i.e., circulating angiogenic cells [CACs] and
mediate intercellular communication of BM-derived
bone marrow [BM]-derived cells) (133). Understand-
cells with other cell types, with modification of the
ing how aging deteriorates stem cell functionality is
transcriptional profile mediated by ncRNAs (147).
of paramount importance to develop new therapeutic
MicroRNA profiling in BM cells from young and aged
approaches in this area.
mice revealed that miR-10A* and miR-21, and their
MECHANISMS OF STEM CELL AGING. Recent data common target gene Hmga2, can drive EPC senes-
obtained in hematopoietic stem cells (HSCs) indicate cence (148). Suppression of those microRNAs in aged
that, by the age of 70 years, clonal diversity collapses, EPCs increased Hmga2 expression and blunted
resulting in the dominance of 1 HSC clone (138). Such expression of the senescence genes p16 INK4a/p19 ARF ,
collapse of highly polyclonal into quasi-monotypic thus ameliorating angiogenic properties. In another
hematopoiesis may influence stem cell competency study, miR-34a induced EPC senescence by sup-
during ischemia or infarction. In addition, several pressing SIRT1 (149). These novel data illustrate how
JACC VOL. 69, NO. 15, 2017 Paneni et al. 1963
APRIL 18, 2017:1952–67 The Aging Cardiovascular System

F I G U R E 3 Features of Age-Related Stem Cell Dysfunction and Approaches to Boost Their Therapeutic Potential in Elderly
Patients With CVD

elderly patient
with CVD (MI, HF)
epigenetic changes
“Dysfunctional
BM-derived cells” genomic instability

clonal diversity

telomere shortening

oxidative stress
“aged” bone marrow
inflammation

ex-vivo reprogramming in situ reprogramming


(epigenetic signals, gene therapy) (cardiomyocyte dedifferentiation,
stimulation of endogenous CSC)

intracoronary stem cell reinfusion

Future strategies to ameliorate the efficiency of autologous transplantation may include ex vivo reprogramming of maladaptive pathways, in
situ reprogramming by stimulating cardiomyocyte dedifferentiation, or in situ stimulation of endogenous cardiac stem cells. BM ¼ bone
marrow; CSC ¼ cardiac stem cell; CVD ¼ cardiovascular disease; HF ¼ heart failure; MI ¼ myocardial infarction.

editing the epigenetic landscape of aged EPCs can the SWISS AMI trial (Swiss Multicenter Intracoronary
furnish a novel mechanism-based approach to pro- Stem Cells Study in Acute Myocardial Infarction),
moting vascular repair in elderly patients with CVD. which recruited a similar number of patients, treat-
ment with BM-derived mononuclear cells for either 5
STEM CELL–BASED THERAPIES. Available evidence to 7 days or 3 to 4 weeks after acute MI failed to
on the CV effects of stem cell-based therapies is improve LV function (assessed by cardiac magnetic
overall very difficult to interpret due to heteroge- resonance imaging) at 12 months (152). Furthermore,
neous populations, and different technical a very recent meta-analysis in patients with re-
approaches. The ACCRUE study (meta-Analysis of fractory angina showed that cell-based therapy im-
Cell-based CaRdiac stUdiEs), which included ran- proves anginal episodes, reduces the use of
domized trials in patients with a recent acute MI, antianginal medications, ameliorates exercise toler-
revealed that intracoronary cell therapy provided no ance and myocardial perfusion, and reduces the risk
benefit, in terms of clinical events or changes in LV of major adverse cardiac events and arrhythmias
function (150). However, the PreSERVE-AMI trial (A compared with maximal medical therapy (153). The
Prospective Randomized Double Blind Placebo explanation behind these conflicting and highly
Controlled Phase II Trial of Intra-coronary Infusion of debated results lies in the fact that, in most cases,
AMR-001, a Bone Marrow Derived Autologous CD34þ “aged” BM cells, which have already traveled down
Selected Cell Product, in Patients With Acute the aforementioned senescence pathways, are rein-
Myocardial Infarction), the largest study of cell-based fused. Moreover, the older recipient organ presents a
therapy for ST-segment elevation MI completed in diseased milieu whereby activation of several patho-
the United States, showed that intracoronary infusion physiological pathways may hamper the healing po-
of autologous CD34 þ cells in patients with ischemic tential of stem cells. In this regard, new-generation
LV dysfunction may exert favorable, albeit small, cell approaches should include identification of compe-
dose-dependent effect on LV ejection fraction, infarct tent cells from the BM aspirate, as well as ex vivo
size, and survival after discharge (151). By contrast, in reprogramming of maladaptive pathways and
1964 Paneni et al. JACC VOL. 69, NO. 15, 2017

The Aging Cardiovascular System APRIL 18, 2017:1952–67

epigenetic signals. Of clinical relevance, the growing JunD, and SIRT1, have gained increasing attention
understanding of chromatin architecture and meta- due to their involvement in essential pathways
bolism has led to the design of specific compounds regulating ROS production and/or scavenging and
(some already approved by the Food and Drug proinflammatory cytokines. Indeed, the SIRT1 acti-
Administration and tested in clinical trials) that can vator SRT2104 has already undergone clinical study
modulate epigenetic modifications, and thereby (157). These more targeted and selective in-
restore gene expression. terventions merit attention, given the disappoint-
ments encountered with the application of blunt
CONCLUSIONS AND FUTURE PERSPECTIVES
approaches to limit oxidative stress in humans.
Recent clinical trials have shown that therapeutic
More than 2 decades of dedicated research have
targeting of oxidative stress using ROS scavengers not
firmly established the concept that increased oxida-
only lacks efficacy, but can even prove harmful (82).
tive stress and inflammation promote CV aging.
Ultimately, from a lifestyle perspective, regimens
However, generalized antioxidant supplementation,
such as caloric restriction or regular endurance exer-
including vitamin E and b-carotene, failed to reduce
cise may attenuate signs of vascular aging by
CV events in asymptomatic individuals, as well as in
increasing SIRT1 levels, PGC-1a –dependent mito-
patients at high CV risk (154,155). Anti-inflammatory
chondrial biogenesis, eNOS functionality, and anti-
interventions, such as anti-TNF a treatments, did
oxidant response via enhanced activity of the
not demonstrate beneficial effects on morbidity
transcription factor Nfr-2 (41,158). Translation of the
and mortality in patients with chronic heart failure
basic and clinical science reviewed here should pre-
(156). Despite these disappointing results, emerging
pare us better to confront the burden of CVD in our
innovative strategies that target ROS, either by
growing older population.
inhibiting specific pro-oxidant enzymes or by aug-
menting endogenous antioxidants, have yielded
some promising preclinical results. Other efforts seek ADDRESS FOR CORRESPONDENCE: Prof. Giovanni G.
to develop selective anti-inflammatory agents that Camici, Center for Molecular Cardiology, University
might reduce CVD without disturbing metabolic ho- of Zurich, Wagistrasse 12, Schlieren CH-8952, Zurich,
meostasis. New molecular targets, such as p66Shc, Switzerland. E-mail: giovanni.camici@uzh.ch.

REFERENCES

1. North BJ, Sinclair DA. The intersection between 8. Newman AB, Naydeck BL, Ives DG, et al. Coro- 15. Chobanian AV. Clinical practice. Isolated sys-
aging and cardiovascular disease. Circ Res 2012; nary artery calcium, carotid artery wall thickness, tolic hypertension in the elderly. N Engl J Med
110:1097–108. and cardiovascular disease outcomes in adults 70 2007;357:789–96.
to 99 years old. Am J Cardiol 2008;101:186–92.
2. Heidenreich PA, Trogdon JG, Khavjou OA, et al. 16. Staessen JA, Gasowski J, Wang JG, et al. Risks
Forecasting the future of cardiovascular disease in 9. Camici GG, Savarese G, Akhmedov A, et al. of untreated and treated isolated systolic hyper-
the United States: a policy statement from the Molecular mechanism of endothelial and vascular tension in the elderly: meta-analysis of outcome
American Heart Association. Circulation 2011;123: aging: implications for cardiovascular disease. Eur trials [Published correction appears in Lancet
933–44. Heart J 2015;36:3392–403. 2001;357:724]. Lancet 2000;355:865–72.
10. Afilalo J, Karunananthan S, Eisenberg MJ, 17. Appel LJ, Espeland MA, Easter L, et al. Effects
3. Fleg JL, Aronow WS, Frishman WH. Cardiovas-
et al. Role of frailty in patients with cardiovascular of reduced sodium intake on hypertension control
cular drug therapy in the elderly: benefits and
disease. Am J Cardiol 2009;103:1616–21. in older individuals: results from the Trial of
challenges. Nat Rev Cardiol 2011;8:13–28.
11. Zieman SJ, Melenovsky V, Kass DA. Mecha- Nonpharmacologic Interventions in the Elderly
4. Christensen K, Doblhammer G, Rau R, et al. (TONE). Arch Intern Med 2001;161:685–93.
nisms, pathophysiology, and therapy of arterial
Ageing populations: the challenges ahead. Lancet
stiffness. Arterioscler Thromb Vasc Biol 2005;25: 18. Cook NR, Appel LJ, Whelton PK. Sodium intake
2009;374:1196–208.
932–43. and all-cause mortality over 20 years in the Trials
5. Freedman VA, Martin LG, Schoeni RF. Recent 12. McEniery CM, Wilkinson IB, Avolio AP. Age, of Hypertension Prevention. J Am Coll Cardiol
trends in disability and functioning among older hypertension and arterial function. Clin Exp Phar- 2016;68:1609–17.
adults in the United States: a systematic review. macol Physiol 2007;34:665–71. 19. Messerli FH, Mancia G, Conti CR, et al. Dogma
JAMA 2002;288:3137–46.
13. Franklin SS, Khan SA, Wong ND, et al. Is pulse disputed: can aggressively lowering blood pres-
6. Kovacic JC, Moreno P, Nabel EG, et al. Cellular pressure useful in predicting risk for coronary sure in hypertensive patients with coronary artery
senescence, vascular disease, and aging: part 2 of heart disease? The Framingham Heart Study. disease be dangerous? Ann Intern Med 2006;144:
a 2-part review: clinical vascular disease in the Circulation 1999;100:354–60. 884–93.
elderly. Circulation 2011;123:1900–10.
14. Franklin SS, Jacobs MJ, Wong ND, et al. Pre- 20. Vidal-Petiot E, Ford I, Greenlaw N, et al. Car-
7. Kuller LH, Lopez OL, Mackey RH, et al. dominance of isolated systolic hypertension diovascular event rates and mortality according to
Subclinical cardiovascular disease and death, among middle-aged and elderly US hypertensives: achieved systolic and diastolic blood pressure in
dementia, and coronary heart disease in pa- analysis based on National Health and Nutrition patients with stable coronary artery disease: an
tients 80þ years. J Am Coll Cardiol 2016;67: Examination Survey (NHANES) III. Hypertension international cohort study. Lancet 2016;388:
1013–22. 2001;37:869–74. 2142–52.
JACC VOL. 69, NO. 15, 2017 Paneni et al. 1965
APRIL 18, 2017:1952–67 The Aging Cardiovascular System

21. Selvaraj S, Steg PG, Elbez Y, et al., REACH 37. Tschudi MR, Barton M, Bersinger NA, et al. the Atherosclerosis Risk in Communities (ARIC)
Registry Investigators. Pulse pressure and risk for Effect of age on kinetics of nitric oxide release in Study. Am J Hypertens 2016;29:114–22.
cardiovascular events in patients with athero- rat aorta and pulmonary artery. J Clin Invest 1996;
53. Fritze O, Romero B, Schleicher M, et al. Age-
thrombosis: from the REACH Registry. J Am Coll 98:899–905.
related changes in the elastic tissue of the human
Cardiol 2016;67:392–403.
38. Taddei S, Virdis A, Ghiadoni L, et al. Age- aorta. J Vasc Res 2012;49:77–86.
22. McEvoy JW, Chen Y, Rawlings A, et al. Dia- related reduction of NO availability and oxidative 54. Liu J, Sukhova GK, Sun JS, et al. Lysosomal
stolic blood pressure, subclinical myocardial stress in humans. Hypertension 2001;38:274–9. cysteine proteases in atherosclerosis. Arterioscler
damage, and cardiac events: implications for blood
39. Lüscher TF, Yang ZH, Diederich D, et al. Thromb Vasc Biol 2004;24:1359–66.
pressure control. J Am Coll Cardiol 2016;68:
Endothelium-derived vasoactive substances: po- 55. Li Z, Froehlich J, Galis ZS, et al. Increased
1713–22.
tential role in hypertension, atherosclerosis, and expression of matrix metalloproteinase-2 in the
23. Beckett NS, Peters R, Fletcher AE, et al., vascular occlusion. J Cardiovasc Pharmacol 1989; thickened intima of aged rats. Hypertension 1999;
HYVET Study Group. Treatment of hypertension in 14 Suppl 6:S63–9. 33:116–23.
patients 80 years of age or older. N Engl J Med
40. Donato AJ, Gano LB, Eskurza I, et al. Vascular 56. Aronson D. Cross-linking of glycated collagen
2008;358:1887–98.
endothelial dysfunction with aging: endothelin-1 in the pathogenesis of arterial and myocardial
24. Williamson JD, Supiano MA, Applegate WB, and endothelial nitric oxide synthase. Am J Phys- stiffening of aging and diabetes. J Hypertens
et al., SPRINT Research Group. Intensive vs stan- iol Heart Circ Physiol 2009;297:H425–32. 2003;21:3–12.
dard blood pressure control and cardiovascular
41. Durrant JR, Seals DR, Connell ML, et al. 57. Amento EP, Ehsani N, Palmer H, et al. Cyto-
disease outcomes in adults aged $75 years: a
Voluntary wheel running restores endothelial kines and growth factors positively and negatively
randomized clinical trial. JAMA 2016;315:2673–82.
function in conduit arteries of old mice: direct regulate interstitial collagen gene expression in
25. Bhatt DL. Troponin and the J-curve of diastolic evidence for reduced oxidative stress, increased human vascular smooth muscle cells. Arterioscler
blood pressure: when lower is not better. J Am superoxide dismutase activity and down- Thromb 1991;11:1223–30.
Coll Cardiol 2016;68:1723–6. regulation of NADPH oxidase. J Physiol 2009;
58. Huang W, Alhenc Gelas F, Osborne-
26. Fuster V. No such thing as ideal blood pres- 587:3271–85.
Pellegrin MJ. Protection of the arterial internal
sure: a case for personalized medicine. J Am Coll
42. Yang YM, Huang A, Kaley G, et al. eNOS elastic lamina by inhibition of the renin-
Cardiol 2016;67:3014–5.
uncoupling and endothelial dysfunction in aged angiotensin system in the rat. Circ Res 1998;82:
27. Forette F, Seux ML, Staessen JA, et al. Pre- vessels. Am J Physiol Heart Circ Physiol 2009;297: 879–90.
vention of dementia in randomised double-blind H1829–36. 
59. Janic M, Lunder M, Sabovic M. Arterial stiff-
placebo-controlled Systolic Hypertension in
43. Santhanam L, Christianson DW, Nyhan D, et al. ness and cardiovascular therapy. Biomed Res Int
Europe (Syst-Eur) trial. Lancet 1998;352:1347–51.
Arginase and vascular aging. J Appl Physiol (1985) 2014;2014:621437.
28. Peila R, White LR, Masaki K, et al. Reducing the 2008;105:1632–42.
60. Walker AE, Henson GD, Reihl KD, et al. Greater
risk of dementia: efficacy of long-term treatment
44. Dikalov SI, Nazarewicz RR. Angiotensin II- impairments in cerebral artery compared with
of hypertension. Stroke 2006;37:1165–70.
induced production of mitochondrial reactive ox- skeletal muscle feed artery endothelial function in
29. Di Bari M, Pahor M, Franse LV, et al. Dementia ygen species: potential mechanisms and relevance a mouse model of increased large artery stiffness.
and disability outcomes in large hypertension tri- for cardiovascular disease. Antioxidants Redox J Physiol 2015;593:1931–43.
als: lessons learned from the systolic hypertension Signaling 2013;19:1085–94. 61. Isabelle M, Simonet S, Ragonnet C, et al.
in the elderly program (SHEP) trial. Am J Epi-
45. Ungvari Z, Orosz Z, Labinskyy N, et al. Chronic reduction of nitric oxide level in adult
demiol 2001;153:72–8.
Increased mitochondrial H2O2 production pro- spontaneously hypertensive rats induces aortic
30. Wilson PW, D’Agostino RB, Levy D, et al. motes endothelial NF-kB activation in aged rat stiffness similar to old spontaneously hypertensive
Prediction of coronary heart disease using risk arteries. Am J Physiol Heart Circ Physiol 2007;293: rats. J Vasc Res 2012;49:309–18.
factor categories. Circulation 1998;97:1837–47. H37–47. 62. Lloyd-Jones DM, Larson MG, Leip EP, et al.
31. Vita JA, Treasure CB, Nabel EG, et al. Coronary 46. Van Guilder GP, Westby CM, Greiner JJ, et al. Lifetime risk for developing congestive heart fail-
vasomotor response to acetylcholine relates to Endothelin-1 vasoconstrictor tone increases with ure: the Framingham Heart Study. Circulation
risk-factors for coronary-artery disease. Circula- age in healthy men but can be reduced by regular 2002;106:3068–72.
tion 1990;81:491–7. aerobic exercise. Hypertension 2007;50:403–9. 63. Redfield MM. Heart failure with preserved
32. Celermajer DS, Sorensen KE, Spiegelhalter DJ, 47. Tang EHC, Vanhoutte PM. Gene expression ejection fraction. N Engl J Med 2016;375:1868–77.
et al. Aging is associated with endothelial changes of prostanoid synthases in endothelial 64. Owan TE, Hodge DO, Herges RM, et al. Trends
dysfunction in healthy men years before the age- cells and prostanoid receptors in vascular smooth in prevalence and outcome of heart failure with
related decline in women. J Am Coll Cardiol muscle cells caused by aging and hypertension. preserved ejection fraction. N Engl J Med 2006;
1994;24:471–6. Physiol Genomics 2008;32:409–18. 355:251–9.
33. Delp MD, Behnke BJ, Spier SA, et al. Ageing
48. Heymes C, Habib A, Yang D, et al. Cyclo- 65. McMurray JJ, Packer M, Desai AS, et al.,
diminishes endothelium-dependent vasodilatation
oxygenase-1 and -2 contribution to endothelial PARADIGM-HF Committees and Investigators.
and tetrahydrobiopterin content in rat skeletal
dysfunction in ageing. Br J Pharmacol 2000;131: Dual angiotensin receptor and neprilysin inhibition
muscle arterioles. J Physiol 2008;586:1161–8.
804–10. as an alternative to angiotensin-converting
34. van der Loo B, Labugger R, Skepper JN, enzyme inhibition in patients with chronic sys-
49. Singh N, Prasad S, Singer D, et al. Ageing is
et al. Enhanced peroxynitrite formation is asso- tolic heart failure: rationale for and design of the
associated with impairment of nitric oxide and
ciated with vascular aging. J Exp Med 2000;192: Prospective comparison of ARNI with ACEI to
prostanoid dilator pathways in the human fore-
1731–44. Determine Impact on Global Mortality and
arm. Clin Sci (Lond) 2002;102:595–600.
morbidity in Heart Failure trial (PARADIGM-HF).
35. Donato AJ, Eskurza I, Silver AE, et al. Direct
50. Küng CF, Lüscher TF. Different mechanisms of Eur J Heart Fail 2013;15:1062–73.
evidence of endothelial oxidative stress with aging
endothelial dysfunction with aging and hyperten-
in humans: relation to impaired endothelium- 66. Otto CM, Prendergast B. Aortic-valve stenosis—
sion in rat aorta. Hypertension 1995;25:194–200.
dependent dilation and upregulation of nuclear from patients at risk to severe valve obstruction.
factor-kB. Circ Res 2007;100:1659–66. 51. Camici GG, Sudano I, Noll G, et al. Molecular N Engl J Med 2014;371:744–56.
pathways of aging and hypertension. Curr Opin
36. Lakatta EG, Levy D. Arterial and cardiac aging: 67. Thanassoulis G, Campbell CY, Owens DS, et al.,
Nephrol Hypertens 2009;18:134–7.
major shareholders in cardiovascular disease en- CHARGE Extracoronary Calcium Working Group.
terprises: part I: aging arteries: a “set up” for 52. Meyer ML, Tanaka H, Palta P, et al. Correlates Genetic associations with valvular calcification and
vascular disease. Circulation 2003;107:139–46. of segmental pulse wave velocity in older adults: aortic stenosis. N Engl J Med 2013;368:503–12.
1966 Paneni et al. JACC VOL. 69, NO. 15, 2017

The Aging Cardiovascular System APRIL 18, 2017:1952–67

68. Libby P. How does lipid lowering prevent response and life span in mammals. Nature 1999; implication of cilostazol after drug-eluting stent
coronary events? New insights from human im- 402:309–13. implantation. J Am Coll Cardiol 2009;53:
aging trials. Eur Heart J 2015;36:472–4. 2298–305.
85. Francia P, delli Gatti C, Bachschmid M, et al.
69. Henein M, Granåsen G, Wiklund U, et al. High Deletion of p66shc gene protects against age- 100. Ota H, Akishita M, Eto M, et al. Sirt1 modu-
dose and long-term statin therapy accelerate related endothelial dysfunction. Circulation lates premature senescence-like phenotype in
coronary artery calcification. Int J Cardiol 2015; 2004;110:2889–95. human endothelial cells. J Mol Cell Cardiol 2007;
184:581–6. 86. Shi Y, Savarese G, Perrone-Filardi P, et al. 43:571–9.

70. Falk RH. Diagnosis and management of the Enhanced age-dependent cerebrovascular 101. Menghini R, Casagrande V, Cardellini M, et al.
cardiac amyloidoses. Circulation 2005;112: dysfunction is mediated by adaptor protein MicroRNA 217 modulates endothelial cell senes-
2047–60. p66Shc. Int J Cardiol 2014;175:446–50. cence via silent information regulator 1. Circula-
87. Spescha RD, Shi Y, Wegener S, et al. Deletion tion 2009;120:1524–32.
71. González-López E, Gallego-Delgado M, Guzzo-
Merello G, et al. Wild-type transthyretin of the ageing gene p66Shc reduces early stroke 102. Zhou S, Chen HZ, Wan YZ, et al. Repression of
amyloidosis as a cause of heart failure with pre- size following ischaemia/reperfusion brain injury. P66Shc expression by SIRT1 contributes to the
served ejection fraction. Eur Heart J 2015;36: Eur Heart J 2013;34:96–103. prevention of hyperglycemia-induced endothelial
2585–94. 88. Spescha RD, Klohs J, Semerano A, et al. Post- dysfunction. Circ Res 2011;109:639–48.
ischaemic silencing of p66Shc reduces ischaemia/ 103. Winnik S, Auwerx J, Sinclair DA, et al.
72. Falk RH. Pondering the prognosis and pathol-
reperfusion brain injury and its expression corre- Protective effects of sirtuins in cardiovascular
ogy of cardiac amyloidosis: answers breed ques-
lates to clinical outcome in stroke. Eur Heart J diseases: from bench to bedside. Eur Heart J 2015;
tions. J Am Coll Cardiol Img 2016;9:139–41.
2015;36:1590–600. 36:3404–12.
73. Rosenberg IH. Sarcopenia: origins and clinical
89. Franzeck FC, Hof D, Spescha RD, et al.
relevance. J Nutr 1997;127:990S–1S. 104. Brunet A, Sweeney LB, Sturgill JF, et al.
Expression of the aging gene p66Shc is increased
Stress-dependent regulation of FOXO transcrip-
74. Curtis E, Litwic A, Cooper C, et al. De- in peripheral blood monocytes of patients with
tion factors by the SIRT1 deacetylase. Science
terminants of muscle and bone aging. J Cell acute coronary syndrome but not with stable
2004;303:2011–5.
Physiol 2015;230:2618–25. coronary artery disease. Atherosclerosis 2012;220:
282–6. 105. Mattagajasingh I, Kim CS, Naqvi A, et al.
75. Reginster JY, Beaudart C, Buckinx F, et al.
SIRT1 promotes endothelium-dependent vascular
Osteoporosis and sarcopenia: two diseases or one? 90. Pagnin E, Fadini G, de Toni R, et al. Diabetes
relaxation by activating endothelial nitric oxide
Curr Opin Clin Nutr Metab Care 2016;19:31–6. induces p66shc gene expression in human pe-
synthase. Proc Natl Acad Sci U S A 2007;104:
ripheral blood mononuclear cells: relationship to
76. Calado RT, Young NS. Telomere diseases. 14855–60.
oxidative stress. J Clin Endocrinol Metab 2005;90:
N Engl J Med 2009;361:2353–65. 106. Martín-Núñez E, Donate-Correa J, Muros-de-
1130–6.
77. Kurz DJ, Kloeckener-Gruissem B, Akhmedov A, Fuentes M, et al. Implications of Klotho in vascular
91. Hernandez JM, Floyd DH, Weilbaecher KN,
et al. Degenerative aortic valve stenosis, but not health and disease. World J Cardiol 2014;6:
et al. Multiple facets of junD gene expression are
coronary disease, is associated with shorter telo- 1262–9.
atypical among AP-1 family members. Oncogene
mere length in the elderly. Arterioscler Thromb 2008;27:4757–67. 107. Ding HY, Ma HX. Significant roles of anti-
Vasc Biol 2006;26:e114–7. aging protein klotho and fibroblast growth fac-
92. Gerald D, Berra E, Frapart YM, et al. JunD re-
78. Brouilette SW, Moore JS, McMahon AD, et al., tor23 in cardiovascular disease. J Geriatr Cardiol
duces tumor angiogenesis by protecting cells from
West of Scotland Coronary Prevention Study 2015;12:439–47.
oxidative stress. Cell 2004;118:781–94.
Group. Telomere length, risk of coronary heart 108. Lindberg K, Olauson H, Amin R, et al. Arterial
93. Paneni F, Osto E, Costantino S, et al. Deletion
disease, and statin treatment in the West of klotho expression and FGF23 effects on vascular
of the activated protein-1 transcription factor
Scotland Primary Prevention Study: a nested case- calcification and function. PLoS One 2013;8:
JunD induces oxidative stress and accelerates age-
control study. Lancet 2007;369:107–14. e60658.
related endothelial dysfunction. Circulation 2013;
79. Zhang W, Chen Y, Wang Y, et al. Short telo- 127:1229–40, e1–21. 109. Kurosu H, Yamamoto M, Clark JD, et al.
mere length in blood leucocytes contributes to the Suppression of aging in mice by the hormone
94. Ricci R, Eriksson U, Oudit GY, et al. Distinct
presence of atherothrombotic stroke and hae- Klotho. Science 2005;309:1829–33.
functions of junD in cardiac hypertrophy and heart
morrhagic stroke and risk of post-stroke death.
failure. Genes Dev 2005;19:208–13. 110. Lord CJ, Ashworth A. The DNA damage
Clin Sci (Lond) 2013;125:27–36.
95. Thompson AM, Wagner R, Rzucidlo EM. Age- response and cancer therapy. Nature 2012;481:
80. Chen S, Lin J, Matsuguchi T, et al. Short 287–94.
related loss of SirT1 expression results in dysre-
leukocyte telomere length predicts incidence and
gulated human vascular smooth muscle cell 111. Vijg J, Suh Y. Genome instability and aging.
progression of carotid atherosclerosis in American
function. Am J Physiol Heart Circ Physiol 2014; Annu Rev Physiol 2013;75:645–68.
Indians: the Strong Heart Family Study. Aging
307:H533–41.
(Albany NY) 2014;6:414–27. 112. Busuttil RA, Dollé M, Campisi J, et al. Genomic
96. Zhang QJ, Wang Z, Chen HZ, et al. Endothelium- instability, aging, and cellular senescence. Ann N Y
81. Haycock PC, Heydon EE, Kaptoge S, et al. specific overexpression of class III deacetylase Acad Sci 2004;1019:245–55.
Leucocyte telomere length and risk of cardiovas- SIRT1 decreases atherosclerosis in apolipoprotein
cular disease: systematic review and meta-analysis. E-deficient mice. Cardiovasc Res 2008;80: 113. Capell BC, Collins FS, Nabel EG. Mechanisms
BMJ 2014;349:g4227. 191–9. of cardiovascular disease in accelerated aging
syndromes. Circ Res 2007;101:13–26.
82. Kornfeld OS, Hwang S, Disatnik MH, et al. 97. Stein S, Lohmann C, Schäfer N, et al. SIRT1
Mitochondrial reactive oxygen species at the heart decreases Lox-1-mediated foam cell formation in 114. Durik M, Kavousi M, van der Pluijm I, et al.
of the matter: new therapeutic approaches for atherogenesis. Eur Heart J 2010;31:2301–9. Nucleotide excision DNA repair is associated with
cardiovascular diseases. Circ Res 2015;116: age-related vascular dysfunction. Circulation
98. Miranda MX, van Tits LJ, Lohmann C, et al.
1783–99. 2012;126:468–78.
The Sirt1 activator SRT3025 provides atheropro-
83. Cosentino F, Francia P, Camici GG, et al. Final tection in Apoe-/- mice by reducing hepatic Pcsk9 115. Shah NR, Mahmoudi M. The role of DNA
common molecular pathways of aging and car- secretion and enhancing Ldlr expression. Eur Heart damage and repair in atherosclerosis: a review.
diovascular disease: role of the p66Shc protein. J 2015;36:51–9. J Mol Cell Cardiol 2015;86:147–57.
Arterioscler Thromb Vasc Biol 2008;28:622–8.
99. Ota H, Eto M, Ako J, et al. Sirolimus and 116. Mahmoudi M, Mercer J, Bennett M. DNA
84. Migliaccio E, Giorgio M, Mele S, et al. The everolimus induce endothelial cellular senescence damage and repair in atherosclerosis. Cardiovasc
p66shc adaptor protein controls oxidative stress via sirtuin 1 down-regulation: therapeutic Res 2006;71:259–68.
JACC VOL. 69, NO. 15, 2017 Paneni et al. 1967
APRIL 18, 2017:1952–67 The Aging Cardiovascular System

117. Botto N, Rizza A, Colombo MG, et al. Evidence 133. Lähteenvuo J, Rosenzweig A. Effects of aging proangiogenic effects and alterations in type 2
for DNA damage in patients with coronary artery on angiogenesis. Circ Res 2012;110:1252–64. diabetics. Blood 2013;121:226–36.
disease. Mutat Res 2001;493:23–30.
134. Zhang H, van Olden C, Sweeney D, et al. 148. Zhu S, Deng S, Ma Q, et al. MicroRNA-10A*
118. Bautista Niño PK, Durik M, Danser AH, et al. Blood vessel repair and regeneration in the and microRNA-21 modulate endothelial progenitor
Phosphodiesterase 1 regulation is a key mecha- ischaemic heart. Open Heart 2014;1:e000016. cell senescence via suppressing high-mobility
nism in vascular aging. Clin Sci (Lond) 2015;129: group A2. Circ Res 2013;112:152–64.
135. Groen BB, Hamer HM, Snijders T, et al. Skel-
1061–75.
etal muscle capillary density and microvascular 149. Zhao T, Li J, Chen AF. MicroRNA-34a in-
119. Jaiswal S, Fontanillas P, Flannick J, et al. Age- function are compromised with aging and type 2 duces endothelial progenitor cell senescence and
related clonal hematopoiesis associated with diabetes. J Appl Physiol (1985) 2014;116: impedes its angiogenesis via suppressing silent
adverse outcomes. N Engl J Med 2014;371:2488–98. 998–1005. information regulator 1. Am J Physiol Endocrinol
Metab 2010;299:E110–6.
120. Costantino S, Paneni F, Cosentino F. Target- 136. Creager MA, Olin JW, Belch JJ, et al. Effect of
ing chromatin remodeling to prevent cardiovas- hypoxia-inducible factor-1a gene therapy on 150. Gyöngyösi M, Wojakowski W,
cular disease in diabetes. Curr Pharm Biotechnol walking performance in patients with intermittent Lemarchand P, et al., ACCRUE Investigators.
2015;16:531–43. claudication. Circulation 2011;124:1765–73. Meta-Analysis of Cell-based CaRdiac stUdiEs
(ACCRUE) in patients with acute myocardial
121. Paneni F, Costantino S, Volpe M, et al. 137. Rajagopalan S, Olin J, Deitcher S, et al. Use of
infarction based on individual patient data. Circ
Epigenetic signatures and vascular risk in type 2 a constitutively active hypoxia-inducible factor-1a
Res 2015;116:1346–60.
diabetes: a clinical perspective. Atherosclerosis transgene as a therapeutic strategy in no-option
2013;230:191–7. critical limb ischemia patients: phase I dose- 151. Quyyumi AA, Vasquez A, Kereiakes D, et al.
escalation experience. Circulation 2007;115: PreSERVE-AMI: a randomized, double-blind, pla-
122. Singhal RP, Mays-Hoopes LL, Eichhorn GL.
1234–43. cebo-controlled clinical trial of intracoronary
DNA methylation in aging of mice. Mech Ageing
administration of autologous CD34þ cells in pa-
Dev 1987;41:199–210. 138. Goodell MA, Rando TA. Stem cells and
tients with left ventricular dysfunction post
healthy aging. Science 2015;350:1199–204.
123. Bollati V, Schwartz J, Wright R, et al. Decline STEMI. Circ Res 2017;120:324–31.
in genomic DNA methylation through aging in a 139. Fadini GP, Losordo D, Dimmeler S. Critical
152. Sürder D, Manka R, Moccetti T, et al. Effect of
cohort of elderly subjects. Mech Ageing Dev reevaluation of endothelial progenitor cell phe-
bone marrow-derived mononuclear cell treat-
2009;130:234–9. notypes for therapeutic and diagnostic use. Circ
ment, early or late after acute myocardial infarc-
Res 2012;110:624–37.
124. Wang JC, Bennett M. Aging and atheroscle- tion: twelve months CMR and long-term clinical
rosis: mechanisms, functional consequences, and 140. Williamson K, Stringer SE, Alexander MY. results. Circ Res 2016;119:481–90.
potential therapeutics for cellular senescence. Circ Endothelial progenitor cells enter the aging arena.
153. Khan AR, Farid TA, Pathan A, et al. Impact of
Res 2012;111:245–59. Front Physiol 2012;3:30.
cell therapy on myocardial perfusion and cardio-
141. Kushner EJ, MacEneaney OJ, Weil BR, et al. vascular outcomes in patients with angina re-
125. Illi B, Ciarapica R, Capogrossi MC. Chromatin
methylation and cardiovascular aging. J Mol Cell Aging is associated with a proapoptotic endothe- fractory to medical therapy: a systematic review
Cardiol 2015;83:21–31. lial progenitor cell phenotype. J Vasc Res 2011;48: and meta-analysis. Circ Res 2016;118:984–93.
408–14.
126. López-Otín C, Blasco MA, Partridge L, et al. 154. Rapola JM, Virtamo J, Ripatti S, et al. Rand-
142. Murasawa S, Llevadot J, Silver M, et al. omised trial of a-tocopherol and b-carotene sup-
The hallmarks of aging. Cell 2013;153:1194–217.
Constitutive human telomerase reverse transcrip- plements on incidence of major coronary events in
127. Brunet A, Berger SL. Epigenetics of aging and tase expression enhances regenerative properties men with previous myocardial infarction. Lancet
aging-related disease. J Gerontol A Biol Sci Med of endothelial progenitor cells. Circulation 2002; 1997;349:1715–20.
Sci 2014;69 Suppl 1:S17–20. 106:1133–9.
155. Jarski RW, Hightower KR, Dangovian MI.
128. Hasegawa Y, Saito T, Ogihara T, et al. 143. He T, Joyner MJ, Katusic ZS. Aging decreases Vitamin E supplementation, cardiovascular events,
Blockade of the nuclear factor-kB pathway in the expression and activity of glutathione peroxidase- and cancer. JAMA 2005;294:425–6; author reply
endothelium prevents insulin resistance and pro- 1 in human endothelial progenitor cells. Microvasc 426.
longs life spans. Circulation 2012;125:1122–33. Res 2009;78:447–52.
156. Coletta AP, Clark AL, Banarjee P, et al. Clinical
129. Schones DE, Leung A, Natarajan R. Chromatin 144. Endtmann C, Ebrahimian T, Czech T, et al. trials update: RENEWAL (RENAISSANCE and
modifications associated with diabetes and obesity. Angiotensin II impairs endothelial progenitor cell RECOVER) and ATTACH. Eur J Heart Fail 2002;4:
Arterioscler Thromb Vasc Biol 2015;35:1557–61. number and function in vitro and in vivo: impli- 559–61.
130. Paneni F, Volpe M, Lüscher TF, et al. SIRT1, cations for vascular regeneration. Hypertension
157. Venkatasubramanian S, Noh RM, Daga S,
p66Shc, and Set7/9 in vascular hyperglycemic 2011;58:394–403.
et al. Cardiovascular effects of a novel SIRT1
memory: bringing all the strands together. Dia- 145. Krankel N, Luscher TF, Landmesser U. Novel activator, SRT2104, in otherwise healthy cigarette
betes 2013;62:1800–7. insights into vascular repair mechanisms. Curr smokers. J Am Heart Assoc 2013;2:e000042.
Pharm Des 2014;20:2430–8.
131. Paneni F, Costantino S, Battista R, et al. 158. Ungvari Z, Parrado-Fernandez C, Csiszar A,
Adverse epigenetic signatures by histone methyl- 146. Paneni F, Costantino S, Kränkel N, et al. et al. Mechanisms underlying caloric restriction
transferase Set7 contribute to vascular dysfunc- Reprogramming ageing and longevity genes re- and lifespan regulation: implications for vascular
tion in patients with type 2 diabetes mellitus. Circ stores paracrine angiogenic properties of early aging. Circ Res 2008;102:519–28.
Cardiovasc Genet 2015;8:150–8. outgrowth cells. Eur Heart J 2016;37:1733–7.

132. Xu S, Yin M, Koroleva M, et al. SIRT6 protects 147. Mocharla P, Briand S, Giannotti G, et al.
against endothelial dysfunction and atheroscle- AngiomiR-126 expression and secretion from KEY WORDS arterial stiffness,
rosis in mice. Aging (Albany NY) 2016;8:1064–82. circulating CD34þand CD14þ PBMCs: role for endothelium, epigenetics, stem cells

You might also like