Download as pdf or txt
Download as pdf or txt
You are on page 1of 22

Aging-Associated

C a rdi o v a s c u l a r
Changes and Their
R e l a t i o n s h i p to
H e a r t F a i l u re
James B. Strait, MD, PhDa,*, Edward G. Lakatta, MDb

KEYWORDS
 Heart failure  Cardiovascular aging  Cardiac function
 Cardiac reserve

Although aging does not itself cause heart failure other cardiovascular disorders, the prevalence of
(HF), it does lower the threshold for manifestation chronic heart failure (CHF) is increasing, with
of the disease. As the populations of most devel- approximately 5.7 million Americans with CHF.4
oped countries continue to become older, on The incidence of HF doubles with each decade
average, the importance of aging as a risk factor of life and the prevalence rises to almost 10% of
for all cardiovascular disease (CVD) increases in those older than 80 years. CHF is a highly lethal
kind (Fig. 1). In the United States alone, it is esti- condition, with significant mortality, morbidity,
mated that there will be 70 million people older and associated costs in the older population.
than 65 by 2030, representing almost 25% of the More than 90% of CHF deaths occur in adults older
population.1 In many respects, HF can be thought than 65 years. CHF is also the leading cause of
of as the quintessential final cardiovascular aging hospitalization in Medicare beneficiaries, with
pathway, representing the convergence of age- those older than 65 accounting for 75% of the
associated changes in cardiovascular structure 1.1 million HF discharge diagnoses.4
and function, aging changes in other organ This article focuses on what is known about
systems, and the progressive increase in cardio- normal cardiovascular aging and the role that
vascular diseases in the elderly. aging-associated changes play in reducing
With the success of treatment options for cardiac reserve to make the heart more suscep-
ischemic and valvular diseases, there is an tible to failure. There are a number of factors that
increasing number of older individuals with some link aging to HF5 and gradually reduce the amount
degree of cardiac damage, type B heart failure at of cardiac reserve until finally the heart is “more
a minimum, who are increasingly imperiled by likely to fail” (Fig. 2).
the diminished cardiac reserve associated with
normal aging. Half of all HF cases are found within 1. Structural changes: There is significant struc-
the 6% of the US population that is older than 752 tural change in the heart and vasculature (eg,
(reviewed by Najjar and colleagues3). These indi- vascular stiffening, increased left ventricular
viduals often go on to develop more severe [LV] wall thickness [within normal limits],
cardiac dysfunction with time. In contrast to and fibrosis) with aging, leading to diastolic
heartfailure.theclinics.com

a
Human Cardiovascular Studies, Laboratory of Cardiovascular Science, Intramural Research Program, Clinical
Research Branch, National Institute on Aging, National Institutes of Health, Harbor Hospital, NM-500, 3001
South Hanover Street, Baltimore, MD 21225, USA
b
Laboratory of Cardiovascular Science, Intramural Research Program, Gerontology Research Center, National
Institute on Aging, National Institutes of Health, 5600 Nathan Shock Drive, Baltimore, MD 21224-6825, USA
* Corresponding author.
E-mail address: Straitj@mail.nih.gov

Heart Failure Clin 8 (2012) 143–164


doi:10.1016/j.hfc.2011.08.011
1551-7136/12/$ – see front matter Published by Elsevier Inc.
144 Strait & Lakatta

relaxation, and sympathetic signaling). These


are described individually in the Cardiac func-
tional, Vascular functional, and Arterial-ventric-
ular interaction sections later in this article.
3. Cardioprotection and repair processes: The
cardiac mechanisms responsible for protection
from injury and injury repair become increas-
ingly defective with age, leading to accentuated
adverse remodeling and increased dysfunction.
4. Increased CVD incidence and prevalence:
Fig. 1. Average prevalence of heart failure according There is a progressive increase in the preva-
to age and sex: Framingham Heart Study, 16-year lence of CVD (eg, coronary artery disease
follow-up. (Data from McKee PA, Castelli WP, McNa- [CAD], hypertension, and diabetes), leading to
mara PM, et al. The natural history of congestive heart the development of ischemic, hypertensive, or
failure: the Framingham study. N Engl J Med
diabetic cardiomyopathy. The reader is referred
1971;285:7796.)
to any number of epidemiologic studies for
evidence of this fact.
5. Systemic disease/Other organ systems: Aging-
dysfunction, increased afterload, and HF with associated changes in other organ systems may
preserved ejection fraction (HFpEF).6,7 These affect cardiac structure-function and thereby
are described individually in the Cardiac struc- contribute to HF development. This facet,
tural and Central Arterial structural sections however, is beyond the scope of the current
later in this article. review.
2. Functional changes: There are functional
changes and compensatory responses that
the aged heart undergoes that diminish its ability
CARDIOVASCULAR CHANGES WITH AGING
to respond to increased workload and decrease
Cardiac Changes at Rest and with Exercise
its reserve capacity (eg, changes in maximal
heart rate, end-systolic volume [ESV], end- Cardiac structural changes
diastolic volume [EDV], contractility, prolonged As described in Table 1 and Fig. 3, there are struc-
systolic contraction, prolonged diastolic tural and functional changes in the heart with

Fig. 2. Pathways linking aging to heart failure. (Modified from Lakatta EG. Age-associated cardiovascular
changes in health: impact on cardiovascular disease in older persons. Heart Failure Rev 2002;7(1):1480; with
permission.)
Aging-Associated Cardiovascular Changes 145

Table 1
Relationship of cardiovascular human aging in health to cardiovascular disease

Possible Relation
Age-Associated Changes Plausible Mechanisms to Human Disease
Cardiovascular structural remodeling
[Vascular intimal thickness [Migration of and [matrix Early stages of atherosclerosis
production by VSMC
Possible derivation of intimal cells
from other sources
[Vascular stiffness Elastin fragmentation Systolic hypertension
[Elastase activity LV wall thickening
[Collagen production by VSMC Stroke
and [ cross-linking of collagen Atherosclerosis
Altered growth factor regulation/
tissue repair mechanisms
[LV wall thickness [LV myocyte size with altered Retarded early diastolic cardiac
Ca21 handling filling
YMyocyte number (necrotic and [Cardiac filling pressure
apoptotic death) Lower threshold for dyspnea
Altered growth factor regulation [Likelihood of heart failure with
Focal matrix collagen deposition relatively normal systolic
function
[Left atrial size [Left atrial pressure/volume [Prevalence of lone atrial
fibrillation and other atrial
arrhythmias
Cardiovascular functional changes
Altered regulation of vascular YNO production/effects Vascular stiffening; hypertension
tone Early atherosclerosis
Reduced threshold for cell Ca21 Changes in gene expression of Lower threshold for atrial and
overload proteins that regulate Ca21 ventricular arrhythmia
handling; increased u6u3 PUFA Increases myocyte death
ration in cardiac membranes Increased fibrosis
[Cardiovascular reserve Lower threshold for, and
increased severity of heart
failure
Reduced physical activity Learned lifestyle Exaggerated age changes in some
aspects of cardiovascular
structure and function.
Negative impact on
atherosclerotic vascular
disease, hypertension, and
heart failure

Abbreviations: LV, Left ventricular; PUFA, polyunsaturated fatty acids.

aging, and each of these can have significant The understanding of the changes in LV mass
implications for CVD. Structurally, there is a signif- with aging has developed over time as researchers
icant increase in myocardial thickness8 as a result have made improvements in exclusion criteria,
of increased cardiomyocyte size.9 In addition, the statistical correction, and technological approach.
heart changes its overall shape from elliptical to The history of this development provides a useful
spheroid, with an asymmetric increase in the insight into the difficulties in conducting aging
interventricular septum more than the free wall.10 research. Initially, autopsy-based studies provided
These changes in thickness and shape have data that suggested cardiac mass increased
important implications for cardiac wall stress and significantly with aging.11 Initial echocardiographic
overall contractile efficiency. studies that calculated LV mass by wall thickness
146 Strait & Lakatta

Fig. 3. Arterial and cardiac changes that occur with aging in healthy humans. LV, left ventricular. (Modified from
Lakatta EG. Cardiovascular regulatory mechanisms in advanced age. Physiol Rev 1993;73:413–65; with permission.)

measurements corroborated these findings; how- and healthy persons of different ages15 has
ever, measurements made from autopsies on repeatedly confirmed the stability of cardiac EF
subjects free from hypertension and CAD then cor- at rest (normal average EF >65%). This is not to
rected for body surface area, suggested that there say that there is no change in components of
is actually no change in the cardiac mass of men cardiac systole. In fact, multiple changes occur
with aging.7 Similarly, autopsies of hospitalized in the mean shortening velocity and the heart’s
patients free of CVD did not show an increase in interactions with the vasculature. The combined
cardiac mass with aging. In fact, they found effects of these individual changes, however,
a decrease in the cardiac mass of men and no balance each other and leave the net systolic func-
change in cardiac mass for women. This finding tion unaltered at rest.
has received support from a magnetic resonance It is with exercise that the effects of aging are
imaging–based study of healthy participants in the most evident (Table 2). An overall decrease in
Baltimore Longitudinal Study of Aging (BLSA),10 exercise tolerance is evident in the progressive
as well as multiple echocardiographic studies.12,13 decline in VO2max, starting at age 20 to 30 and
Based on these and other studies, it now appears falling by approximately 10% per decade
that there is no change in LV mass in women and (Fig. 4A, B).16 Additionally, the rate of this decline
an actual decrease in LV mass in men with aging. progressively increases with age. The meaning of
The increased wall thickness represents an asym- these changes and insight into the underlying
metric increase in the interventricular septum factors can be clarified through a review of the
more than the free wall redistributing cardiac Fick equation:
muscle, but not increasing total cardiac mass.
VO2 max 5 CO  ðA  VÞO2

Cardiac Functional Changes where CO 5 cardiac output, AO2 5 arterial O2


Despite the aging-associated changes that may content, and VO2 5 venous O2 content.
limit a person’s functional capacity and promote Cross-sectional data reveal that the VO2max
vascular stiffening with consequent increased falls an average of 50% from age 20 to 80 overall,
afterload, the overall resting systolic function of which must then be a result of either/both CO and
cardiac muscle does not change (see Fig. 3) (A – V)O2. CO is known to fall 25% with aging,
with healthy aging. Examination of echocardio- which by definition must be because of changes
graphic LV shortening fraction14 and radionuclide in stroke volume (SV) or heart rate (HR).17 Because
ejection fraction (EF) in normotensive subjects SV is thought to be maintained throughout life (our
Aging-Associated Cardiovascular Changes 147

Table 2
laboratory is currently completing studies to test
Exhaustive upright exercise: changes in aerobic this directly), the bulk of the CO decline is likely
capacity and cardiac regulation between the because of impaired heart-rate acceleration (or
ages of 20 and 80 years in healthy men and maximal heart rate). Although the LV-EDV
women increases modestly with exercise, this results in
an overall maintenance of SV (although our labora-
Oxygen consumption Y(50%) tory has ongoing studies to assess this assertion
(A-V)O2 Y(25%) directly). The remaining 25% change in VO2max
Cardiac index Y(25%) is a result of a change in the (A – V)O2 term, reflect-
Heart rate Y(25%) ing changes in oxygen extraction efficiency and
Stroke volume No D
balance (determinants include muscle mass, mito-
chondrial efficiency, the ability to redistribute
Preload
blood flow to working muscles, and so forth18,19)
EDV [(30%) (men > women) and suggests the importance of muscle mass,
Afterload mitochondrial efficiency, and so forth. Put another
Vascular (PVR) [(30%) way, the decline in VO2max is largely a result of
Cardiac (ESV) [(275%) changes in the oxygen pulse, which is the product
LC contractility Y(60%) of SV and the difference between peak VO2 and
Ejection fraction Y(15%) arterial oxygen concentration or peak VO2/
Plasma [ maximal HR. Overall, this reduction in cardiac
catecholamines reserve is a result of multiple factors, including
Cardiac and vascular Y increased vascular afterload, arterial-ventricular
responses to load mismatching, reduced intrinsic myocardial
b-adrenergic contractility, impaired autonomic regulation, and
stimulation physical deconditioning.

Abbreviations: (A-V)O2, arterial-venous oxygen concen- Cardiac Diastolic Function


tration; EDV, end diastolic volume; ESV, end systolic
volume; LV, left ventricular; PVT, peripheral vascular Despite maintenance of systolic function at rest,
resistance. there are a number of changes in the diastolic
phase of the cardiac cycle that occur with aging.

Fig. 4. Longitudinal changes in peak VO2 by gender, predicted from the mixed-effects model, and separated by
gender in panel A with the percent change by decade represented in panel B. (A) Peak VO2 declines progressively
more steeply with advancing age, with similar declines in men and women. Note that peak VO2 is only slightly
higher in men than women at younger ages, converging by old age. (B) Per-decade longitudinal changes in
peak VO2 by gender and age decade, derived from the mixed-effects model. Note that longitudinal declines
in peak VO2 steepen with age and that men decompensate at an accelerated, although similar rate after age
60. (Modified by Ferrucci L from Fleg JL, Morrell CH, Bos AG, et al. Accelerated longitudinal decline of aerobic
capacity in healthy older adults. Circulation 2005;112:674–82; with permission).
148 Strait & Lakatta

Normal diastolic filling can be divided into 2 pattern of relaxation.10,22,23 Surprisingly, although
phases: passive (represented by the “E wave” on the rate of filling declines and is shifted to later in
echocardiographic study of transmitral flow) and diastole, the EDV actually remains unchanged
active (represented by an A wave and produced or increases somewhat with rest and low-level
by atrial contraction) (Fig. 5A). The heart fills with exercise, largely as a result of the slower24 heart
blood more slowly in older versus younger healthy rate, which permits longer filling time (see Fig. 3,
individuals, resulting in a lower proportion of total Table 2) and increase in the ESV. Taken together,
diastolic filling occurring during this passive, early these reductions in early diastolic filling are par-
diastolic phase,8,20,21 mainly because of an tially compensated for by changes in adrenergic
increase in the isovolumic relaxation time. As the signaling that lower the maximal heart rate;
bulk of ventricular filling shifts to later in diastole however, these compensations are not sufficient
and there is significant atrial enlargement with to maintain cardiac functional reserve when
aging, the atrium contributes a greater portion of a subject is exposed to maximal exercise, as
the total EDV and a decrease in the E/A ratio shown in Fig. 5C. In fact, a significant deficit is
(see Fig. 5B). uncovered in the older population when the peak-
Normal exercise induces an increase in SV and filling rate at maximal exercise is compared in
heart rate to increase overall cardiac output (see young versus older individuals.
Table 2). This increased heart rate also increases
the rate of isovolumic relaxation and produces Changes in cardiac conduction system and
a “suction” effect, which helps fill the ventricle. electrocardiogram
These responses are diminished with aging, Aging is associated with a generalized increase in
however, as a result of slowed relaxation, reduced elastic and collagenous tissue. Fat accumulates
b-adrenergic responsiveness, and alterations in the around the sinoatrial node, sometimes creating

Fig. 5. Diastolic function at rest and with exercise. (A) The Doppler diastolic time-flow-velocity profile, showing
the E and A waves from which the indexes of diastolic filling performance are derived. Time is represented on the
horizontal axis. A simultaneous ECG is shown as a timing reference to indicate atrial and ventricular activation. LV
indicates left ventricular. (B) The ratio of early left ventricular diastolic filling rate (E) to the atrial filling compo-
nent (A) declines with aging, and the extent of this E/A decline with aging in healthy BLSA volunteers is identical
to that in participants of the Framingham Study. (C) Maximum LV filling rate at rest and during vigorous cycle
exercise assessed via equilibrium gated blood-pool scans in healthy volunteers from the BLSA. EDV, end-
diastolic volume. ([B, C] From Strait JS, Lakatta EG. Cardiac aging: aging from human to molecules. Chapter 44.
Muscle 2011; with permission.)
Aging-Associated Cardiovascular Changes 149

a partial or complete separation of the node from In addition, there are increases in atrial arrhyth-
the atrial tissue. A pronounced decline in the mias, atrial fibrillation, paroxysmal supraventric-
number of pacemaker cells occurs after age 60; ular tachycardia, and ventricular arrhythmias with
by age 75, fewer than 10% of the number seen aging.14 The prevalence of these arrhythmias, as
in young adults remain. A variable degree of calci- well as their association with mortality, are detailed
fication on the left side of the cardiac skeleton also in Table 4. Atrial fibrillation (AF) is found in approx-
occurs. This can affect the atrioventricular node, imately 3% to 4% of subjects older than 60, a rate
atrioventricular bifurcation, and proximal left and 10-fold higher than the general adult population.
right bundle branches leading to significant risk Short bursts of paroxysmal supraventricular
for atrioventricular conduction block. tachycardia (PSVT) on a resting electrocardiogram
Although there is generally no change in the (ECG) are found in 1% to 2% of healthy individuals
resting heart rate with aging, a number of changes older than 65. Twenty-four-hour ambulatory moni-
do occur in the cardiac conduction system that toring studies have demonstrated short runs of
affect its electrical properties (Table 3). A review PSVT (usually 3 to 5 beats) in 13% to 50% of clin-
by Fleg and Lakatta14 provides greater detail of ically healthy older persons.30,31 The incidence
this area. Likely because of a decrease in parasym- increases with exercise, from 0% in the 20s to
pathetic activity, there is a reduction in the level of approximately 10% in the 80s. Ventricular ectopic
phasic variation in R-R interval with respiration,25 beats (VEBs) undergo an exponential increase in
as well as the occurrence of sinus bradycardia. prevalence with advancing age. Pooled data
There can be an increase in pathologic sinus brady- from nearly 2500 ECGs from hospitalized patients
cardia owing to sick sinus syndrome and conduc- older than 70 revealed VEBs in 8%.32
tion pathway, although the heart rate is stable in The available data in older adults without ap-
the healthy aged heart. The P-R interval, represent- parent heart disease support a marked age-related
ing atrioventricular conduction, increases from 159 increase in the prevalence and complexity of
ms at ages 20 to 35, to 172 ms beyond age 60.26 exercise-related VEBs, at least in men; however,
The QRS axis shifts leftward, possibly because of the prognosis conferred by frequent or repetitive
increases in LV wall thickness, with 20% of healthy VEBs induced by exercise in such individuals is
subjects having a left axis deviation by age 100.27 unclear.
Interestingly, despite increased LV thickness, there In summary, aging is associated with multiple
is a decline in the R-wave and S-wave amplitudes ECG changes in persons without evidence of
with aging evident by age 40.28 There is an increase CVD. Such changes include a blunted respiratory
in nonspecific ST-T changes with aging, although
the relation of these to clinical heart disease
remains in question.29 Table 4
Relationship of arrhythmias to age and
mortalitya

Effect of
Table 3
Age on Effect on
Normal age-associated changes in resting
Arrhythmia Prevalence Mortality
electrocardiogram measurements
Supraventricular Increased None
Effect on ectopic beats
Measurement Change with Age Mortality Paroxysmal Increased Probably none
R-R interval No change supraventricular
tachycardia
P-wave duration Minor increase None
Atrial fibrillation Increased Increased
P-R interval Increase None
(chronic)
QRS duration No change
Ventricular ectopic Increased Probably none
QRS axis Leftward shift None beats
QRS voltage Decrease None Ventricular Increased Probably none
Q-T interval Minor increase Probable tachycardia
increase (short runs)
T-wave voltage Decrease None
a
In healthy elderly.
Modified from Lakatta EG, Sollott SJ, Pepe S. The old Modified from Lakatta EG, Sollott SJ, Pepe S. The old
heart: operating on the edge. Novartis Found Symp heart: operating on the edge. Novartis Found Symp
2001;235:172–96 [discussion: 196–201, 217–20]; with 2001;235:172–96 [discussion: 196–201, 217–20]; with
permission. permission.
150 Strait & Lakatta

sinus arrhythmia; a mild P–R interval prolongation; Na1/Ca21 exchanger. Studies in rodents that
a leftward shift of the QRS axis; and increased have used gene therapy or exercise conditioning
prevalence, density, and complexity of ectopic to increase the level of SERCA2 have demon-
beats, both atrial and ventricular. Although these strated that impaired relaxation and Ca21 seques-
findings generally do not affect prognosis in tration that occur with aging can be reduced by
clinically healthy older adults, other findings these interventions. Finally, there is a compensa-
that become more prevalent with age, such as tory increase in L-type Ca21 currents with an
increased QRS voltage, Q waves, QT interval increase in the apparent number and activity of
prolongation, and ST–T-wave abnormalities, are individual L-type calcium channels that prolongs
generally associated with increased cardiovas- the action potential, a slower inactivation of the
cular risk. Abnormalities, such as left bundle L-type channel, and a reduction in outwardly
branch block or AF, are strongly predictive of directed K1 currents, all of which serve to prolong
future cardiac morbidity and mortality among older the action potential.
adults, even if asymptomatic. Ca21 regulation is significantly affected by reac-
tive oxygen species (ROS) and is therefore involved
Changes in cellular Ca21 handling in complex pathways (see Fig. 6) involving
Arrhythmias are a by-product of aberrant Ca21 membrane polyunsaturated fatty acids (PUFAs).
handling. Maintenance of the calcium-electro- PUFAs are an important family of dietary fats
chemical gradient is an intricate process for the (including the u-3 and u-6 PUFAs) that undergo
individual cardiomyocyte, as depicted in Fig. 6 lipid peroxidation to produce aldehydes, akenals,
and Table 1. With aging, several changes occur and hydroxyalkenals, such as 4-hydroxy-2-nonenal
(Table 5) that slow the cellular reactions control- (HNE). HNE reacts with protein sulfhydryl groups
ling the beat of the heart as a whole (more details to induce altered protein conformations and inflict
can be found in Lakatta and Sollott33 and Janc- damage on the cell.33 With advancing age, a
zewski and colleagues34). The action potential, number of changes occur in the lipid content of
transient increase in cytosolic Ca21, and rate of the cell membrane that reduce its capacity
contraction are all prolonged, which consequently to tolerate and adapt to stress (eg, ischemia-reper-
prolong systole and diastole of the heart,33,35 fusion) (see Fig. 7).
consistent with the lower maximal heart rate Notable among these changes are a relative
seen in older individuals during exercise. This is decrease in the u-3:u-6 PUFA content ratio in
because of changes in several ion currents, as cellular membranes, enhanced cellular production
well as a reduced rate of Ca21 reuptake through of ROS, and alterations in proteins governing Ca21
downregulation of SERCA2 protein levels, mobilization, especially the decreased expression
decreased phospholamban phosphorylation, and or function of SERCA2 and Na1-Ca21 exchange
an approximately 50% increase in levels of the protein.33,36 These changes lead to disturbances

Fig. 6. An overview of change in the aging heart that predisposes to a reduced threshold for abnormal Ca21
handling during acute stress. See text for details. (Adapted from Lakatta EG, Sollott SJ, Pepe S. The old heart:
operating on the edge Novartis Found Symp 2001;235:172–96 [discussion: 196–201, 217–20]; with permission.)
Aging-Associated Cardiovascular Changes 151

Table 5
Myocardial changes with adult aging in rodents

Ionic, Biophysical/Biochemical
Functional Changes Mechanisms Molecular Mechanisms
21
Prolonged contraction Prolonged cytosolic Ca
transient
YSR Ca21 pumping rate YSR Ca21 pump mRNA (no D
calsequestrin mRNA)
YPump site density
Prolonged action potential YIca Inactivation [Na/Ca exchanger mRNA
YITo density
Diminished contraction velocity YaMHC protein YaMHC mRNA
[bMHC protein [b MHC mRNA (no D actin mRNA)
YMyosin ATPase activity YRxRb1 and Y mRNA
YRxRb1 and Y mRNA
YRxRb1 and Y protein
YThyroid receptor protein
Diminished b-adrenergic YCoupling BAR-ACyclase (no D Gi YbiAR mRNA (no D BARK mRNA)
contractile response activation, no D BARK activity)
YTNI phospholamban
phosphorylation
YIca augmentation
YCai transient augmentation
[Enkephalin peptides
[Proenkephalin peptides
Myocardial stiffness [Hydroxyline proline content [Collagen mRNA
[Activity of myocardia RAS [Fibronectin mRNA
[AT1R mRNA
[Atrial naturetic peptide [Atrial naturetic peptide mRNA
Growth response YInduction of immediate early
genes
YCardiomyocyte proliferation D in CyclinD1, D2, D3, pRb, p130,
CDK-2
YCardiomyocyte survival D in TERT, IGF-1, caspases, AIF,
survivin
Heart shock response YActivation of HSF

Abbreviations: AIF, Apoptosis Inducing Factor; BARK, Beta Adrenergic Receptor Kinase; CDK, Cyclin Dependent Kinase;
HSF, Heat Shock Factor; IGF-1, insulinlike growth factor 1; RAS, renin-angiotensin system; TERT, telomerase reverse tran-
scriptase; TNI, Troponin I.

in excitation-contraction coupling and in intracel- mitochondrial permeability transition, which


lular Ca21 compartmentalization, leading in turn involves the release of mitochondrial contents
to spontaneous Ca21 oscillations and arrhythmias and activation of a sequence of events that lead
(see Fig. 6). In addition, mitochondrial dysfunction to cell death. Recent research has demonstrated
can ensue, which impairs energy metabolism that these effects of aging are exacerbated by
(decreased ATP production) and increases ROS poor nutritional habits, but can be ameliorated by
production. Overproduction of ROS and the diets substituting u-3 PUFAs for u-6 PUFAs.
inability to scavenge excess ROS lead to
damaging lipid peroxidation and relatively more Cardiac adrenergic responsiveness
diffusible but still potent, reactive intermediates, Adrenergic signaling is an important component of
such as HNE, which affect widespread protein aging-associated cardiovascular change. Acute
targets and amplify Ca21 dysregulation and mito- exercise and other stressors stimulate sympa-
chondrial abnormalities. Finally, the abnormal thetic modulation of the cardiovascular system,
Ca21 handling and ROS buildup can induce which increases heart rate, augments myocardial
152 Strait & Lakatta

Fig. 7. Changes in the injury and protection thresholds in aging heart. (A) Aging diminishes the heart’s threshold
to sustain injury (eg, from ischemia/reperfusion). Lifestyle modifications, including exercise and possibly caloric
restriction, may partially diminish the aging effect. Comorbidities (such as diabetes) have negative influence.
(B) Aging increases the heart’s threshold to activate protection-signaling mechanisms. Various pharmacologic
agents (eg, sulfonylureas, antioxidants, partial fatty acid oxidation inhibitors, and cyclooxygenase-2 inhibitors)
that can interfere with cardioprotective signaling pathways can exacerbate this trend and further increase the
protection threshold. Exercise and caloric restriction might attenuate the age-dependent trends. (Modified
from Juhaszova M, Rabuel C, Zorov DB, et al. Protection in the aged heart: preventing the heart-break of old
age? Cardiovasc Res 2005;66:233–44; with permission.)

contractility and relaxation, reduces LV afterload, from the supine basal state, plasma levels of norepi-
and redistributes blood to working muscles and nephrine (NE) and epinephrine increase to a greater
skin to dissipate heat. With aging, however, there extent in older than in younger healthy individ-
is a diminishment of the autonomic modulation of uals.37,38 This increase in plasma catecholamines
heart rate, LV contractility, and arterial afterload,37 appears to be a compensatory response to the
related to a decline in the efficiency of postsyn- reduced cardiac muscarinic b-receptor density
aptic b-adrenergic signaling. There is a decrease and functional decline with advancing age. The
in cardiovascular responses to b-adrenergic amount of circulating NE rises, in part because of
antagonist infusion at rest with aging (Fig. 8A) increased spillover from tissues (including the
and a similarity in the hemodynamic profile of heart37–39) as a result of deficient NE reuptake at
younger b-blocked subjects to older unblocked nerve endings during acute exercise, as well as
individuals (see Fig. 8B, C) that is consistent reduced plasma clearance. With prolonged exer-
with this mechanistic explanation. Some smaller cise, this diminished neurotransmitter reuptake
studies37 provide further evidence for this effect. can result in depletion of stores and reduced release
For example, significant b-blockade–induced LV and spillover in the body, thereby contributing
dilation occurs only in younger subjects, the heart further to the blunted cardio-acceleration and LV
rate is reduced to a greater degree by acute b- systolic performance seen with age during such
adrenergic blockade in younger versus older exercise.
subjects, and the age-associated deficits in LV
early diastolic filling rate, both at rest and during Renin-angiotensin system in cardiac tissue
exercise, can be mimicked by b-adrenergic The mechanism behind age-related cardiac re-
blockade. modeling is not completely understood, although
The mechanism behind these changes is not a number of clues have been found6,33: (1) there
entirely clear, although several alterations in the is a strong body of evidence for the central role
adrenergic system have been observed. It is note- for the renin-angiotensin system (RAS); (2)
worthy that the apparent deficits in sympathetic increased oxidative stress has been implicated in
modulation of cardiac and arterial functions with age-related remodeling; (3) aging has been associ-
aging occur in the presence of elevated sympathetic ated with increased production of ROS in several
neurotransmitter levels: during any perturbation studies; (4) RAS activation itself may stimulate
Aging-Associated Cardiovascular Changes 153

Fig. 8. (A) Stroke volume index (SVI) as a function of end-diastolic volume index (EDVI) at rest (R) and during
graded cycle workloads in the upright-seated position in healthy Baltimore Longitudinal Study of Aging
(BLSA) men in the presence and absence (dashed lines) of b-adrenergic blockade. R, seated rest; 1–4 or 5, graded
submaximal workloads on cycle ergometer; max, maximum effort. Stroke volume versus end-diastolic functions
with symbols are those measured in the presence of propranolol; dashed line functions without symbols are the
stroke volume as versus end-diastolic functions measured in the absence of propranolol. Note that in the absence
of propranolol, the SVI versus EDVI relation in older persons (dashed lines) is shifted rightward from that in
younger persons (dashed lines with points). This indicates that the left ventricle (LV) of older persons in the sitting
position compared with that of younger persons operates from a greater preload both at rest and during
submaximal and maximal exercise. Propranolol markedly shifts the SV-EDVI relationship in younger persons
(triangle without points) rightward, but does not markedly offset the curve in older persons (circle). Thus,
with respect to this assessment of ventricular function curve, b-adrenergic blockade with propranolol makes
younger men appear like older men. The abolition of the age-associated differences in the LV function curve after
propranolol are accompanied by a reduction in heart rate, which at maximum, is shown in (B). (B) Peak exercise
heart rate in the same subjects as in (A) in the presence and absence of acute b-adrenergic blockade by propran-
olol. (C) The age-associated reduction in peak LV diastolic filling rate at maximal exercise in healthy BLSA subjects
is abolished during exercise in the presence of b-adrenergic blockade with propranolol. Y, <40 years; O, >60 years.
* Represents P<.0001 (From Strait JS, Lakatta EG. Cardiac aging: aging from human to molecules. Chapter 44.
Muscle 2011; with permission.)

ROS production; and (5) nicotinamide adenine by the aging vasculature40 and overall decrease in
dinucleotide phosphate (NADPH) oxidases appear cardiomyocyte number. This initiates growth factor
to be important in linking these processes. signaling (eg, angiotensin II [ANGII]/transforming
One important stimulus for RAS signaling is growth factor beta [TGF-b]), which has been shown
thought to be the stretch of cardiac myocytes and in vitro to promote cell growth and matrix produc-
fibroblasts owing to the increased load imposed tion, as well as increasing apoptosis.41 This ANGII
154 Strait & Lakatta

does not likely come from circulating stores but is flow, blood vessels are dynamic structures that
instead released from fibroblasts and cardiomyo- adapt, repair, remodel, and govern their structural
cytes in an autocrine/paracrine manner in response and function properties using complex signaling
to stretch.42 ANGII binds to both angiotensin I (AT1) pathways in response to load, stress, and age.
and angiotensin II (AT2) receptors to activate
a complex signal transduction cascade that affects
a number of transcriptionally important factors.43 Central arterial structural changes
Either directly or through AT1, ANGII can activate Macroscopic structural changes A number of age-
the NADPH oxidase complex to generate ROS. associated structural changes occur in the arterial
Understanding of the relative importance of these system, including thickening and dilation of large
numerous pathways will be an important compo- arteries (see Fig. 3, Table 1).44 Echocardiographic
nent of cardiac aging research and will provide an studies show that the aortic root dilates modestly
important link to similar signaling changes in the with age, approximating 6% between the fourth
arterial wall. and eighth decades (Fig. 9A). Similarly, serial
chest x-rays over 17 years have demonstrated
that the aortic knob diameter increases from 3.4
Central Arterial Changes with Aging
to 3.8 cm, although there are data to suggest
In studying the effects of aging on the cardiovascular that in hypertensive individuals (when aortic diam-
system, it is important to recall its anatomic and eter is corrected for covariates), it may represent
functional position in series with the vascular a relative decrease in effective diameter that may
system. In fact, many researchers now believe that contribute to increased load on the heart.45 In
the greatest risk factor for development of CVD is normal aging, however, such aortic root dilation
“unsuccessful” age-associated arterial aging. provides an additional stimulus for LV hypertrophy
Rather than acting as simple conduits for blood because the larger volume of blood in the proximal

Fig. 9. Age-associated changes in arterial structure and function in men (stars) and women (squares) in the BLSA.
Best-fit regression lines (quadratic or linear) are shown for men (solid lines) and women (dotted lines). (A) Aortic
root size (measured by M-mode echocardiography) indexed to body surface area. (B) Common carotid-intima-
medial thickness (measured by B-mode ultrasonography). (C) Carotid-femoral pulse wave velocity (an index of
central arterial stiffness). (From Najjar SS, Lakatta EG, Gerstenblith G. Cardiovascular aging: the next frontier in
cardiovascular prevention. In: Blumenthal R, Foody J, Wong NA, editors. Prevention of cardiovascular disease:
companion to Braunwald’s heart disease. Philadelphia: Saunders; 2011. p. 415–32; with permission.)
Aging-Associated Cardiovascular Changes 155

aorta leads to a greater inertial load against which (AT1). All of these components have been found to
the senescent heart must pump. increase within the aged arterial wall in various
Autopsy reports published as early as 1910 species.54,55 The local ANGII concentration is
described age-associated aortic thickening. Cross- more than 1000-fold that of circulating ANGII, is
sectional studies using ultrasound imaging have independently regulated, and plays an important
demonstrated the intimal-medial layer thickens role in vascular pathophysiology with aging. ANGII
nearly threefold (see Fig. 9B) between the ages of protein abundance increases in the aged aortic
20 and 90 years in apparently healthy individuals.46 wall in rats. Studies of nonhuman primates also
Both the average and range of intimal-medial thick- show that ANGII, ACE, and chymase, elements of
ness measurements is greater at higher ages, the classic RAS, are all upregulated in the aged arte-
suggesting a variable response to chronologic age rial wall.55,56 The marked increase of ANGII in the
that merits further study to identify the components aged arterial wall appears to offset the effect of
of “successful aging.” reduced plasma levels of ANGII in the elderly. In
addition, the molecules linked to the ANGII sig-
Microscopic Structural and Biochemical Changes naling cascade, including calpain-1, matrix metallo-
Aging is associated with structural and functional proteinase types 2 and 9 (MMP-2 and MMP-9),
changes of the arterial wall media (hypertrophy, monocyte chemotactic protein-1 (MCP-1), and
extracellular matrix accumulation, calcium deposits) transforming growth factor-beta 1 (TGF-b1) are up-
and the vascular endothelium (decrease in the regulated within the aged arterial wall (see Tables 1
release of vasodilators and increased synthesis of and 3).54,57,58 A number of studies support the
vasoconstrictors) that are associated with increased concept that ANGII signaling is a central pathway
vascular stiffness (see Table 1).47,48 Collagen and that mediates the cellular and molecular mecha-
elastin provide the strength and elasticity, respec- nisms that underlie arterial aging.55
tively, of the arterial wall and are normally stabilized
by enzymatic cross-linking. With aging, an increase Central arterial function
in collagen content, nonenzymatic collagen cross- Blood pressure Arterial pressure is determined by
linking, and fraying of elastin fibrils occur in the the interplay of peripheral vascular resistance
medial layer,49 all of which reduce arterial distensi- (PVR) and arterial stiffness. Stiffness, as used
bility and increase stiffness. The occurrence of here, refers to time-varying cardiac elastance
irreversible nonenzymatic glycation-based cross- throughout the cardiac cycle as a result of
linking of collagen to form advanced glycation end combined effects of active contractile and
products increases with age and is associated with “passive” structural properties, as well as the
increased arterial stiffness in elderly people.50 interaction of the two. Systolic blood pressure
Furthermore, receptors for advanced glycation end (SBP), which is influenced by arterial stiffness,
products are stimulated to produce inflammatory PVR, and cardiac function, rises with age even in
and stress responses. normotensive cohorts. In contrast, diastolic blood
Through its secretion of nitric oxide (NO) and pressure (DBP), rises with increased PVR but is
endothelin, the endothelium is a powerful regulator lowered by arterial stiffness, resulting in an
of arterial tone. Endothelial dysfunction has been increase in diastolic pressure until age 50,
identified in several cardiovascular disorders, a leveling off between 50 and 60, then a decline
including hypertension, hypercholesterolemia, and after age 60.59 Thus, hypertension in the elderly
coronary and peripheral atherosclerosis.51 Human is often characterized by isolated or predominant
and animal studies have revealed that aging SBP elevation. Pulse pressure, the difference
is associated with a reduction in endothelial- between SBP and DBP, is a useful clinical index
dependent vasodilatation, thought secondary to of arterial stiffness and the pulsatile load on the
reduced NO bioavailability.49 A number of animal arterial tree and typically increases with aging.
studies have found that NO production and NO Some studies have suggested that pulse pressure
levels52 decline with aging, likely as a result of is a more powerful predictor of future cardiovas-
a decline in the level of endothelial nitric oxide syn- cular events than either SBP or DBP in older
thase (eNOS).53 adults.60
In addition to the structural alterations (described
previously), arterial function is governed by age- Pulse wave velocity and reflected pulse
associated changes in several signaling cascades, waves Central arterial stiffening occurs with aging
most prominently the RAS. The classic RAS is even in the absence of clinical hypertension,61 as
composed of angiotensinogen, renin, angiotensin measured by pulse wave velocity (PWV). The
(ANG) I and II, angiotensin-converting enzyme Augmentation Index (AI) illustrates an important
(ACE), chymase, angiotensin, and ANGII receptor site of cardiac-vascular interaction. When the
156 Strait & Lakatta

forward pulse wave reaches an area of impedance the arterial tree, typically from the carotid region to
mismatch (vessel bifurcation or movement to the femoral artery. Multiple studies have shown
a higher resistance vessel), a reflected wave that that aortofemoral PWV increases with age, typi-
travels back up the arterial tree toward the central cally twofold to threefold across the adult lifespan
aorta is generated. This reflected wave is identified (see Fig. 9C).64 Furthermore, PWV has been
as a small notch, inflection point, in the carotid and shown, both in clinically healthy cohorts and those
radial pulse waveforms, measured by arterial ap- with CV disease, to be a predictor of future cardio-
planation tonometry. The situation with AI is a bit vascular events, independent of blood pressure.65
more complicated (not shown). It increases until
about age 50,62 even in clinically healthy volun-
Arterial-Ventricular Coupling
teers. The clinical significance of these AI changes
with age is that in young subjects, the reflected Arterial ventricular coupling background
wave typically arrives back at the proximal aorta The concept of coupling between effective arterial
in diastole and may assist in coronary artery dia- elastance (EA) and ventricular elastance measures
stolic filling. In older individuals, the reflected (ELV) is exceedingly important to the study of
waves travel faster, thus arriving at the proximal cardiovascular aging, and so merits further discus-
aorta during late systole, thereby creating an sion (Fig. 10).66 Oftentimes, a by-product of the
increased load for the ventricle, a failure to need to reduce the cardiovascular system to
augment DBP, and a potential compromise of a manageable model system is an excessive focus
coronary blood flow. Several studies in cohorts on individual components rather than consider-
with CVD have observed that higher AI is associ- ation of their function and interaction with the
ated with adverse clinical outcomes.63 A second entire system. The ratio of effective EA/ELV is an
method of arterial stiffness assessment, PWV, is index of arterial ventricular coupling, related to
a Doppler-based method that measures the speed the inverse of EF, and provides a more accurate
with which an arterial pressure wave travels along measure of the complex cardiovascular system.

Fig. 10. Ventricular pressure-volume diagram from which effective arterial elastance (EA) and LV end-systolic ela-
stance (ELV) are derived. EA represents the negative slope of the line joining the end-diastolic volume (EDV) and
the end-systolic pressure (ESP) points. ELV represents the slope of the end-systolic pressure-volume relationship
passing through the volume intercept (VO). The shaded area represents the cardiac stroke work (SW), and the
hatched area represents the potential energy (PE). LV ESP is the LV pressure at the end of systole. EDV is the LV
volume at the end of diastole. End-systolic volume (ESV) is the LV volume at the end of systole. Stroke volume
(SV) is the volume of blood ejected by the LV with each beat and is obtained from subtracting ESV from EDV. BP,
blood pressure; EF, ejection fraction; PVA, pressure-volume area. (From Chantler PD, Lakatta EG, Najjar SS.
Arterial-ventricular coupling: mechanistic insights into cardiovascular performance at rest and during exercise. J
Appl Physiol 2008;105:1342–51; with permission; and Sunagawa K, Maughan WL, Burkhoff D, et al. Left ventricular
interaction with arterial load studied in isolated canine ventricle. Am J Physiol 1983;245(5 Pt 1):H773–80; with
permission.)
Aging-Associated Cardiovascular Changes 157

The effective EA is a steady-state arterial param- Arterial-ventricular coupling at rest


eter that characterizes the functional properties of As detailed previously, most of the components of
the arterial system by incorporating peripheral EA change with age (eg, SBP, pulse pressure), so
vascular resistance, total lumped vascular compli- the fact that the EA/ELV ratio is maintained at rest
ance, characteristic impedance, and systolic and (Fig. 11A–C), despite the known changes in the
diastolic time intervals (characterized by the rela- vascular system and consequent increases in
tionship of end systolic pressure and stroke afterload, suggests important adaptive changes
volume). The ELV (left ventricular end-systolic ela- in the cardiac muscle itself.66 These strategies (in
stance) represents myocardial performance and part) appear to involve LV wall thickening and
is defined by the relationship of end-systolic pres- increased end-systolic pressure8 (moderates
sure to end systolic volume. LV wall tension), increased end-systolic volume,

Fig. 11. (A) Arterial ventricular coupling indexed for body surface area (EAI/ELVI), (B) Effective arterial elastance
indexed to body surface area (EAI). (C) Effective LV elastance indexed to body surface area (ELVI) in men (dashed
lines) younger than 40 years (triangle) and older than 60 years (circle), as well as women (solid lines) younger
than 40 years (diamond) and older than 60 years (star) in the supine and seated positions, at 50% of maximal work-
load, and at peak exercise. EAI/ELVI decreases during exercise in both young and older men and women (P<.0001);
however, older men and women have a blunted decline in EAI/ELVI (P<.001). EAI increases during exercise in both
young and older men and women (P<.0001). At maximal exercise, EAI is greater in older versus younger women
(P<.002). In contrast, EAI does not differ between young and older men. ELVI increases during exercise in both young
and older men and women (P<.0001). At maximal exercise, ELVI is greater in younger versus older men (P<.001) and
tended to be greater in younger than older women (P <.07). (From Strait JS, Lakatta EG. Cardiac aging: aging from
human to molecules. Chapter 44. Muscle 2011; with permission; and Modified from Najjar SS, Schulman SP, Fleg JL,
et al. Relationship of age and sex on ventricular-vascular coupling at rest and exercise. Circulation 2000;102(Suppl);
II–602; with permission.)
158 Strait & Lakatta

prolongation of systolic contraction time67 (main- markedly change in healthy subjects.73 Thus, the
tains normal ejection time in the presence of late limited capacity of patients with systolic HF
augmentation of aortic impedance), cardiac to augment their cardiovascular function during
shape,68 and aspects of the diastolic phase. times of stress (such as exercise) involve marked
In HF, these relationships change. In systolic deficits in both LV and arterial elastance reserves.
HF, patients have a downward and rightward shift To date, there are no studies that have examined
of the end-systolic P-V relationship and thus the change in EA/ELV or EA during exercise in HFpEF.
a reduced ELVI (left ventricular end-systolic Researchers have examined the change in ELV and
elastance index) (range 0.6–2.6 mm Hg/mL1/ EF in HFpEF during upright bicycle exercise.74
m2).69 Patients with systolic HF have an augmented Compared with hypertensive controls with LV
EAI (range 1.7–3.7 mm Hg/mL1/m2) owing to hypertrophy, patients with HFpEF had a threefold
a decrease in stroke volume index (SVI) and to smaller increase in ELV and a reduced ability to lower
increases in HR and PVR.69 The increase in EAI their PVR and increase their HR during exercise.
and decrease in ELVI result in marked, up to threefold They also had a 50% smaller increase in EF during
to fourfold, increases in EA/ELV (range 1.3–4.3). This exercise. As EF is inversely related to EA/ELV, this
suboptimal coupling reflects diminished cardiovas- suggests that the change in EA/ELV during exercise
cular performance and efficiency of the failing heart. in HFpEF may also be severely blunted. Because
In contrast, patients who have HF with preserved female sex and systolic hypertension are risk
ejection fraction (HFpEF) have an 18% and 20% factors for HFpEF, and because the pathophysi-
higher resting EA and ELV, respectively, compared ology of HFpEF involves a limited cardiovascular
with healthy controls.70 Studies suggest a matched reserve,75 the diminished EA/ELV reserve observed
increase in EA and ELV and therefore no difference in in systolic hypertensive women suggests that they
their EA/ELV when compared with normotensive may be exhibiting signs of subclinical (Stage B)
controls or hypertensive patients without HF. HF. This raises the possibility that they may be on
a trajectory to progressive exercise intolerance
Arterial-ventricular coupling with exercise and perhaps functional limitations.
Arterial-ventricular coupling with exercise has
significant implications for an individual’s exercise Cardioprotective and Repair Processes
capacity with aging, however.66 Otherwise normal
arteries dilate and stiffen with age, imposing Because of the many factors discussed previously,
increased load in the form of inertance, reflected the aged heart is placed under increasing levels
pulse waves, artery compliance, and resistance (re- of stress owing to its diminished functional
viewed by Lakatta71). This increased load has reserve. Furthermore, increasing age results in
a significant impact on the relationship of the heart the increased occurrence of numerous disease
to the vascular system, as represented by the EA/ processes (eg, diabetes, hypertension). Although
ELV ratio and described previously. Furthermore, it the heart has protective systems in place to deal
increases tremendously when the body exercises with such insult (see Fig. 2), these decline with
(see Fig. 11). Although the resting EA/ELV ratio is age (see Fig. 7A), resulting in a lower injury
preserved by the parallel increase in both variables threshold. It is not yet clear if there are interventions
(described previously), exercise requires an (eg, physical conditioning) that can be instituted in
increase in EF that the aged heart may have diffi- the elderly to delay or reverse these processes, but
culty providing. To increase EF, the ELV must there are a number of promising candidates.
increase to a greater extent than the EA. With Physical conditioning
increasing age, however, ELV fails to increase in There is much evidence that aerobic exercise
proportion to the increased EA in men and there is programs can provide improvements in peak
a deficit in LVEF reserve when older subjects are oxygen consumption and its components, as well
exposed to acutely increased workloads (ie, exer- as increases in ventilatory threshold and submax-
cise). Interestingly, reduction of the afterload by an imal endurance for older persons with HF.76 A
acute infusion of sodium nitroprusside in healthy, longitudinal study in older men studied in the
older BLSA volunteers augments LVEF at rest and upright position indicates that an enhanced phys-
with exercise and provides further evidence of the ical conditioning status increases O2 consumption
importance of vascular properties.72 and work capacity, in part by increases in the
The baseline alterations in EA/ELV and its maximum CO by increasing the maximum SV,
components in patients with systolic HF are also and in part by increasing the estimated total body
evident during exercise.73 Indeed, EA, ELV, and (A-V)O2 use.15 The augmentation of maximum SVI
EA/ELV do not appreciably change during exercise is a result of an augmented reduction of LVESV15
in patients with systolic HF, whereas they and, thus, a concomitant increase in LVEF, as the
Aging-Associated Cardiovascular Changes 159

effect of conditioning status to increase Left telomere-related proteins.79,80 The telomeres of


Ventricular End Diastolic Volume Index (ml/m2) newly generated cells in senescent human hearts
(LVEDVI) exercise is minimal (recall that LVEDVI quickly shorten to lengths similar to the aged car-
during acute vigorous exercise is already appre- diomyocytes they are replacing. Telomeres nor-
ciably increased in older, sedentary, precondi- mally serve as protective caps for chromosomal
tioned men77). This minor effect of physical ends, protecting them from DNA damage repair
conditioning on maximum exercise LVEDVI in older systems and activation of the p16INK4a pathway,
persons is in contrast to the effect of physical with eventual senescence or apoptosis.81
conditioning in younger persons, which substan- However, telomeres lose 30 to 150 base pairs
tially increases LVEDVI and SVI during vigorous during each cell division owing to the inability of
exercise on the basis of the Frank Starling mecha- DNA polymerase to fully replicate the 3’ end of
nism, as well as via an enhanced LVEF. In contrast the DNA strand. When the telomere becomes
to the improved LV ejection, the maximum heart “too short,” the cell can no longer divide, and it
rate of older persons does not vary with physical becomes senescent and eventually dies. Telo-
conditioning status and exercise does not appear meres can be extended by telomerase, which is
to offset the age-associated deficiency in sympa- composed of 2 main components, the telomerase
thetic modulation. It is noteworthy that the RNA component (TERC) and telomerase reverse
increased pulse wave velocity, carotid augmenta- transcriptase (TERT), with a third component (dys-
tion index, or pulse pressure that occurs with aging kerin) working to stabilize the complex. Agonism of
is less in older persons who are physically condi- telomerase activity through these components
tioned than in sedentary persons.59 An effect of may have some benefit in stabilizing cellular DNA.
physical conditioning to reduce these components If the promise of EPC mobilization in the aged
of vascular afterload appears to be involved in the heart is to be realized, the problems of telomere
effect of conditioning to improve LV ejection.61 shortening and cellular “memory” will have to be
dealt with, as well as methods to control the
Stem cells and the promise of cardiac “internal clock” that resides in DNA-telomeres.5 In-
regeneration sulinlike growth factor (IGF-1) also holds promise as
It had traditionally been thought that cardiomyo- a regulator of cardiac senescence. Activation of the
cytes terminally differentiate and withdraw from IGF-1/IGF-1R pathway preserves telomere length
the cell cycle early in their development; however, and promotes cardiac progenitor cell growth and
carbon dating methods have established that car- survival.82 Injection of IGF-1 in damaged myocar-
diomyocytes continue to be synthesized and a low dium promotes the migration and homing of cardiac
level of turnover in the heart persists throughout stem cells and facilitates neovascularization.83
life.78 New cells can arise from endothelial
Ischemic preconditioning
precursor cells (EPCs) that exist in the bone
Ischemic preconditioning (IPC) is the heart’s
marrow or precursor cells (PCs) from other loca-
endogenous capacity to resist ischemic damage
tions, including adipose tissue and the heart itself.
and its effects, including suppression of ventricular
There has been great interest in using resident
arrhythmias and enhanced recovery of contractile
EPCs as a source of myocardial regeneration after
function.84 The process is dependent on an initial,
injury or other degradatory processes and many
brief ischemic event usually lasting fewer than 5
studies of cardiac cell turnover.
minutes. It appears to have both an early protective
The field remains contentious. Some researchers
function lasting about 1 hour after preconditioning,
contend that the rate of cardiomyocyte turnover
and also a late-delayed action that returns approx-
gradually decreases with aging from 1% per year
imately 24 to 96 hours later. Unfortunately, aging
for a 25-year-old to 0.45% per year for a 75-year-
results in decreased effectiveness of this and other
old.78 Alternatively, Anversa and colleagues79 found
protective pathways,84 which leads to adecreased
that cardiomyocyte regeneration from predomi-
injury threshold in the aged heart (see Fig. 7A, B).
nantly cardiac resident stem cells actually increases
This is but one example of a system that could be
with aging and remains important in the heart
manipulated to enhance the cardiac protective
throughout life. These newly generated cells appear
systems in the aging heart (see Fig. 7B).
to quickly degrade in older individuals, however, to
resemble those they have replaced. Necrotic cellular
CLINICAL APPROACH TO HEART FAILURE IN
destruction progresses with aging until the heart is
AGED PERSONS
unable to keep pace with the level of required stem
Clinical Assessment
cell replacement, leading to a decline in cell numbers.
The cause of this “aging memory” may be Evaluation of the older patient presenting with
reduced telomerase activity, ROS, or loss of failure symptoms should include a noninvasive
160 Strait & Lakatta

study to determine whether the primary problem is individuals with HF and preserved systolic
impaired systolic function. Although systolic function.
dysfunction is present in at least half of CHF cases, ACE inhibitors are a cornerstone of therapy in
the presence of a normal, or elevated, EF is more patients with systolic dysfunction and their benefit
common in older than younger patients with HF, extends to the elderly. Their benefit in patients with
particularly women and those with atrial fibrillation preserved systolic function, however, is less
and hypertension. The number of patients with certain.87 There is an ongoing trial of spironolac-
heart failure and preserved systolic function is tone in patients with HF with preserved systolic
increasing, and is likely to continue to increase function, but first outcomes are not expected
with the aging of the general population. before 2011. There are no clear explanations for
It is often helpful to investigate whether any the lack of survival benefit for any medical inter-
reversible precipitants are present in older individ- vention in older patients with HF with preserved
uals who present with new-onset or worsening HF systolic function. The diagnosis is more difficult
symptoms, including anemia, infection, thyroid and thus patients without heart disease, for
disease, atrial fibrillation, and dietary or medica- whom these therapies would not be expected to
tion noncompliance. Investigation for common co- be of any benefit, may be included in some of
morbidities is also useful, as they are frequent and the studies. The pathophysiology may differ, with
associated with increased hospitalizations and myocardial fibrosis and impaired ventricular-
adverse clinical outcomes.85 Hypertension is vascular coupling more important responsible
almost invariably present, as well as increased mechanisms, and these may not respond to the
central vascular stiffness and therefore impaired tested therapies. Treatment might, therefore, be
ventricular-vascular coupling. It should be noted focused on blood pressure control in those with
that evidence of long-standing volume overload hypertension, rate control in those with atrial fibril-
and an S3 gallop is less likely in patients with lation, and judicious use of diuretics in the setting
preserved systolic function and that in the general of volume overload. It should be noted, however,
older population with HF, exertional symptoms are that the steep volume/pressure relationship asso-
less common, and those related to fatigue and ciated with increased vascular stiffness in the
mental status changes more common. The diag- older individual, might result in symptomatic
nosis of HF with preserved systolic function is hypotension when diuretics result in intravascular
primarily one of exclusion in patients with objective volume depletion.
evidence of pulmonary vascular congestion Devices (eg, implantable cardiac defibrillators,
without findings of an ischemic, hypertensive, or biventricular pacing devices, left ventricular assist
valvular etiology. Amyloid should also be consid- devices) may also significantly improve outcomes
ered in older patients presenting with HF symp- in patients with persistent failure despite medical
toms in the absence of other identifiable causes. therapy. There has been significant development
on this front that has made their benefits acces-
sible to patients throughout the spectrum of
Considerations with Pharmacologic
disease; however, as with any treatment, there is
Interventions
no guarantee of benefit and it is often just as help-
Diuretics are particularly useful in older patients ful to first assess more socially relevant factors.
with increased vascular stiffness presenting with This is especially relevant in the elderly, who
acute congestive symptoms, as significant reduc- have a diminished cardiac reserve to begin with
tions in pressure occur with relatively small and may see significantly different treatment bene-
changes in intravascular volume. HF with primary fits when compared with the typically younger
systolic dysfunction and with preserved systolic study population on which many therapeutic
function are both associated with increased recommendations are based. The importance of
sympathetic and renin-angiotensin-aldosterone the individual patient’s role as a partner in his or
activation. In those with primary systolic dysfunc- her care and of individualizing treatment and moni-
tion, b-blockers improve survival and other impor- toring plans cannot be overemphasized.
tant cardiovascular outcomes in the older subsets Although patients may carry the same HF diag-
of large randomized trials.86 It is unfortunate that nosis, they differ markedly in terms of disease
many older individuals with HF and systolic severity and complexity, associated comorbid-
dysfunction are not receiving any b-blocker ities, social support, education, ingrained habits,
therapy, or are underdosed. In contrast to the access to medical personnel and knowledge,
many successful trials in those with systolic and understanding of health care information and
dysfunction, there are no reported randomized directions. Noncompliance with medications or
studies limited to the effects of b-blockers in older diet is often cited as a major factor contributing
Aging-Associated Cardiovascular Changes 161

to hospitalization in patients with HF. In the older Lung Transplantation. Circulation 2009;119(14):
patient, however, noncompliance is more likely 1977–2016.
because of social isolation, financial difficulties, 5. Chen W, Frangogiannis NG. The role of inflamma-
limited travel and meal options, decreased tolera- tory and fibrogenic pathways in heart failure asso-
bility for some medicines, comorbidities, and ciated with aging. Heart Fail Rev 2010;15(5):
difficult-to-follow complex medical regimens. In 415–22, 1–8.
many of these patients, a multidisciplinary team 6. Lakatta EG, Levy D. Arterial and cardiac aging:
approach, including simplification of the medical major shareholders in cardiovascular disease enter-
regimen, close monitoring, and intensive patient prises: part II: the aging heart in health: links to heart
education, can decrease hospital admission and disease. Circulation 2003;107(2):346–54.
improve quality of life. 7. Scholz DG, Kitzman DW, Hagen PT, et al. Age-
related changes in normal human hearts during
the first 10 decades of life. Part I (Growth): a quanti-
SUMMARY
tative anatomic study of 200 specimens from
Aging results in an increase in CVD and a decrease subjects from birth to 19 years old. Mayo Clin Proc
in cardiac reserve at the same time that the repair 1988;63(2):126–36.
processes designed to deal with these problems 8. Gerstenblith G, Frederiksen J, Yin F, et al. Echocar-
become less active/effective. These factors diographic assessment of a normal adult aging pop-
combine to set the stage for heart failure: (1) struc- ulation. Circulation 1977;56(2):273–8.
turally, the heart thickens and stiffens with age, 9. Olivetti G, Melissari M, Capasso JM, et al. Cardiomy-
resulting in the increased imposition of a number opathy of the aging human heart. Myocyte loss and
of functional demands; (2) functionally, changes reactive cellular hypertrophy. Circ Res 1991;68(6):
that assist the resting heart to deal with the effects 1560–8.
of aging cause significant functional deficits with 10. Hees PS, Fleg JL, Lakatta EG, et al. Left ventricular
exercise or stress, thereby lowering the cardiac remodeling with age in normal men versus women:
reserve that the younger heart can call on to deal novel insights using three-dimensional magnetic reso-
with disease or insult; and (3) finally, although the nance imaging. Am J Cardiol 2002;90(11):1231–6.
increased incidence of disease, less structurally 11. Linzbach AJ, Akuamoa-Boateng E. [Changes in the
efficient heart, and decreased cardiac reserve aging human heart. I. Heart weight in the aged]. Klin
associated with aging would be well served by Wochenschr 1973;51(4):156–63 [in German].
an effective repair system, this too declines with 12. Khouri MG, Maurer MS, El-Khoury Rumbarger L.
age. It is hoped that improved understanding of Assessment of age-related changes in left ventric-
the aged heart will enable the development of ther- ular structure and function by freehand three-
apies that prevent the genesis of HF or at dimensional echocardiography. Am J Geriatr Cardiol
a minimum help clinicians to treat the unique prop- 2005;14(3):118–25.
erties of the failing, senescent heart. 13. Dannenberg AL, Levy D, Garrison RJ. Impact of age
on echocardiographic left ventricular mass in
REFERENCES a healthy population (the Framingham Study). Am
J Cardiol 1989;64(16):1066–8.
1. Writing Group Members, Lloyd-Jones D, Adams RJ, 14. Fleg JL, Lakatta EG. Normal Aging of the Cardiovas-
et al. Heart disease and stroke statistics—2010 cular System. Chapter in Cardiovascular Disease in
update: a report from the American Heart Associa- the Elderly. 4th edition. In: Aronow W, Rich MW,
tion. Circulation 2010;121(7):e46–215. Fleg JL, editors. Boca Raton (FL): CRC Press;
2. Thomas S, Rich M. Epidemiology, pathophysiology, 2008. p. 1–43.
and prognosis of heart failure in the elderly. Heart 15. Schulman SP, Lakatta EG, Fleg JL, et al. Age-related
Fail Clin 2007;3(4):381–7. decline in left ventricular filling at rest and exercise.
3. Najjar SS, Gerstenblith G, Lakatta EG. Aging and the Am J Physiol 1992;263(6 Pt 2):H1932–8.
Cardiovascular System. Cardiovascular Medicine. In: 16. Fleg JL, Lakatta EG. Role of muscle loss in the age-
Willerson JT, Wellens HJ, Cohn JN, et al, editors. Lon- associated reduction in VO2 max. J Appl Physiol
don: Springer London; 2007. p. 2439–51. 1988;65(3):1147–51.
4. Jessup M, Abraham WT, Casey DE, et al. 2009 17. Fleg JL, Morrell CH, Bos AG, et al. Accelerated
focused update: ACCF/AHA Guidelines for the longitudinal decline of aerobic capacity in healthy
Diagnosis and Management of Heart Failure in older adults. Circulation 2005;112(5):674–82.
Adults: a report of the American College of Cardi- 18. Le Blanc PR, Rakusan K. Effects of age and
ology Foundation/American Heart Association Task isoproterenol on the cardiac output and regional
Force on Practice Guidelines: developed in collabo- blood flow in the rat. Can J Cardiol 1987;3(5):
ration with the International Society for Heart and 246–50.
162 Strait & Lakatta

19. Rakusan K, Blahitka J. Cardiac output distribution in rats is an adaptation to sustain youthful intracellular
rats measured by injection of radioactive microspheres Ca21 regulation. J Mol Cell Cardiol 2002;34(6):
via cardiac puncture. Can J Physiol Pharmacol 1974; 641–8.
52(2):230–5. 35. Lakatta E. Age-associated cardiovascular changes
20. Miyatake K, Okamoto M, Kinoshita N, et al. Augmen- in health: impact on cardiovascular disease in older
tation of atrial contribution to left ventricular inflow persons. Heart Fail Rev 2002;7(1):29–49.
with aging as assessed by intracardiac Doppler 36. Lakatta EG, Sollott SJ, Pepe S. The old heart: oper-
flowmetry. Am J Cardiol 1984;53(4):586–9. ating on the edge. Novartis FoundSymp 2001;235:
21. Bryg RJ, Williams GA, Labovitz AJ. Effect of aging 172–96 [discussion: 196–201, 217–20].
on left ventricular diastolic filling in normal subjects. 37. Lakatta EG. Cardiovascular regulatory mechanisms
Am J Cardiol 1987;59(9):971–4. in advanced age. Physiol Rev 1993;73(2):413–67.
22. Spina RJ, Turner MJ, Ehsani AA. Beta-adrenergic- 38. Lakatta EG. Deficient neuroendocrine regulation of
mediated improvement in left ventricular function the cardiovascular system with advancing age in
by exercise training in older men. Am J Physiol healthy humans. Circulation 1993;87(2):631–6.
1998;274(2 Pt 2):H397–404. 39. Esler MD, Turner AG, Kaye DM, et al. Aging effects
23. Tan Y, Wenzelburger F, Lee E, et al. The pathophys- on human sympathetic neuronal function. Am J
iology of heart failure with normal ejection fraction: Physiol 1995;268(1 Pt 2):R278–85.
exercise echocardiography reveals complex abnor- 40. Lakatta E. Cardiovascular aging research: the next
malities of both systolic and diastolic ventricular horizons. J Am Geriatr Soc 1999;47(5):613–25.
function involving torsion, untwist, and longitudinal 41. Cigola E, Kajstura J, Li B, et al. Angiotensin II acti-
motion. J Am Coll Cardiol 2009;54(1):36. vates programmed myocyte cell death in vitro. Exp
24. Tarasov KV, Sanna S, Scuteri A, et al. COL4A1 is Cell Res 1997;231(2):363–71.
associated with arterial stiffness by genome-wide 42. Sadoshima J, Xu Y, Slayter HS, et al. Autocrine
association scan. Circ Cardiovasc Genet 2009; release of angiotensin II mediates stretch-induced
2(2):151–8. hypertrophy of cardiac myocytes in vitro. Cell
25. Hiss RG, Lamb LE. Electrocardiographic findings in 1993;75(5):977–84.
122,043 individuals. Circulation 1962;25:947–61. 43. Cave AC, Brewer AC, Narayanapanicker A, et al.
26. Fleg JL, Das DN, Wright J, et al. Age-associated NADPH oxidases in cardiovascular health and
changes in the components of atrioventricular disease. Antioxid Redox Signal 2006;8(5–6):691–728.
conduction in apparently healthy volunteers. 44. Lakatta EG, Wang M, Najjar SS. Arterial aging and
J Gerontol 1990;45(3):M95–100. subclinical arterial disease are fundamentally
27. Golden GS, Golden LH. The “Nona” electrocardio- intertwined at macroscopic and molecular levels.
gram: findings in 100 patients of the 90 plus age Med Clin North Am 2009;93(3):583–604, Table of
group. J Am Geriatr Soc 1974;22(7):329–32. Contents.
28. Harlan WR, Graybiel A, Mitchell RE, et al. Serial elec- 45. Farasat SM, Morrell CH, Scuteri A, et al. Pulse pres-
trocardiograms: their reliability and prognostic valid- sure is inversely related to aortic root diameter
ity during a 24-yr. period. J Chronic Dis 1967;20(11): implications for the pathogenesis of systolic hyper-
853–67. tension. Hypertension 2008;51(2):196–202.
29. Mihalick MJ, Fisch C. Electrocardiographic findings 46. Nagai Y, Metter E, Earley C, et al. Increased carotid
in the aged. Am Heart J 1974;87(1):117–28. artery intimal-medial thickness in asymptomatic old-
30. Manolio TA, Furberg CD, Rautaharju PM, et al. Cardiac er subjects with exercise-induced myocardial
arrhythmias on 24-h ambulatory electrocardiography ischemia. Circulation 1998;98(15):1504–9.
in older women and men: the Cardiovascular Health 47. Ungvari Z, Kaley G, de Cabo R, et al. Mechanisms
Study. J Am Coll Cardiol 1994;23(4):916–25. of vascular aging: new perspectives. J Gerontol A
31. Fleg JL, Kennedy HL. Cardiac arrhythmias in Biol Sci Med Sci 2010;65(10):1028–41.
a healthy elderly population: detection by 24-hour 48. Zieman SJ, Melenovsky V, Kass DA. Mechanisms,
ambulatory electrocardiography. Chest 1982;81(3): pathophysiology, and therapy of arterial stiffness.
302–7. Arterioscler Thromb Vasc Biol 2005;25(5):932–43.
32. Fisch C. Introduction: chronic ventricular arrhyth- 49. Lakatta EG, Levy D. Arterial and cardiac aging:
mias—a major unresolved health problem. Heart major shareholders in cardiovascular disease enter-
Lung 1981;10(3):451–4. prises: part I: aging arteries: a “set up” for vascular
33. Lakatta EG, Sollott SJ. Perspectives on mammalian disease. Circulation 2003;107(1):139–46.
cardiovascular aging: humans to molecules. Comp 50. Semba RD, Najjar SS, Sun K, et al. Serum
Biochem Physiol A Mol Integr Physiol 2002;132(4): carboxymethyl-lysine, an advanced glycation end
699–721. product, is associated with increased aortic pulse
34. Janczewski AM, Spurgeon HA, Lakatta EG. Action wave velocity in adults. Am J Hypertens 2009;
potential prolongation in cardiac myocytes of old 22(1):74–9.
Aging-Associated Cardiovascular Changes 163

51. Csiszar A, Wang M, Lakatta EG, et al. Inflammation 66. Chantler PD, Lakatta EG, Najjar SS. Arterial-
and endothelial dysfunction during aging: role of ventricular coupling: mechanistic insights
NF-kappaB. J Appl Physiol 2008;105(4):1333–41. into cardiovascular performance at rest and during
52. Tschudi MR, Barton M, Bersinger NA, et al. Effect of exercise. J Appl Physiol 2008;105(4):1342–51.
age on kinetics of nitric oxide release in rat aorta and 67. Lakatta EG. Cardiac muscle changes in senes-
pulmonary artery. J Clin Invest 1996;98(4):899–905. cence. Annu Rev Physiol 1987;49:519–31.
53. Cernadas MR, Sánchez de Miguel L, Garcı́a- 68. Hees PS, Fleg JL, Mirza ZA, et al. Effects of normal
Durán M, et al. Expression of constitutive and induc- aging on left ventricular lusitropic, inotropic, and
ible nitric oxide synthases in the vascular wall of chronotropic responses to dobutamine. J Am Coll
young and aging rats. Circ Res 1998;83(3):279–86. Cardiol 2006;47(7):1440–7.
54. Wang M, Takagi G, Asai K, et al. Aging increases 69. Asanoi H, Sasayama S, Kameyama T. Ventriculoar-
aortic MMP-2 activity and angiotensin II in nonhuman terial coupling in normal and failing heart in humans.
primates. Hypertension 2003;41(6):1308–16. Circ Res 1989;65(2):483–93.
55. Wang M, Monticone RE, Lakatta EG. Arterial aging: 70. Lam CSP, Roger VL, Rodeheffer RJ, et al. Cardiac
a journey into subclinical arterial disease. Curr structure and ventricular-vascular function in
Opin Nephrol Hypertens 2010;19(2):201–7. persons with heart failure and preserved ejection
56. Jiang L, Wang M, Zhang J, et al. Increased aortic fraction from Olmsted County, Minnesota. Circula-
calpain-1 activity mediates age-associated angio- tion 2007;115(15):1982–90.
tensin II signaling of vascular smooth muscle cells. 71. Lakatta EG. Arterial aging is risky. J Appl Physiol
PLoS One 2008;3(5):e2231. 2008;105(4):1321–2.
57. Wang M, Zhao D, Spinetti G, et al. Matrix metallopro- 72. Nussbacher A, Gerstenblith G, O’Connor FC, et al.
teinase 2 activation of transforming growth factor- Hemodynamic effects of unloading the old heart.
beta1 (TGF-beta1) and TGF-beta1-type II receptor Am J Physiol 1999;277(5 Pt 2):H1863–71.
signaling within the aged arterial wall. Arterioscler 73. Cohen-Solal A, Faraggi M, Czitrom D, et al. Left
Thromb Vasc Biol 2006;26(7):1503–9. ventricular-arterial system coupling at peak exercise
58. Wang M, Zhang J, Walker SJ, et al. Involvement of in dilated nonischemic cardiomyopathy. Chest 1998;
NADPH oxidase in age-associated cardiac remodel- 113(4):870–7.
ing. J Mol Cell Cardiol 2010;48(4):765–72. 74. Borlaug BA, Melenovsky V, Russell SD, et al.
59. Tanaka H, Dinenno FA, Monahan KD, et al. Aging, Impaired chronotropic and vasodilator reserves limit
habitual exercise, and dynamic arterial compliance. exercise capacity in patients with heart failure and
Circulation 2000;102(11):1270–5. a preserved ejection fraction. Circulation 2006;
60. Roman MJ, Devereux RB, Kizer JR, et al. High 114(20):2138–47.
central pulse pressure is independently associ- 75. Kitzman DW, Higginbotham MB, Cobb FR, et al.
ated with adverse cardiovascular outcome the Exercise intolerance in patients with heart failure
strong heart study. J Am Coll Cardiol 2009; and preserved left ventricular systolic function:
54(18):1730–4. failure of the Frank-Starling mechanism. J Am Coll
61. Vaitkevicius PV, Fleg JL, Engel JH, et al. Effects of Cardiol 1991;17(5):1065–72.
age and aerobic capacity on arterial stiffness in 76. Fleg JL. Can exercise conditioning be effective in
healthy adults. Circulation 1993;88(4 Pt 1):1456–62. older heart failure patients? Heart Fail Rev 2002;
62. Mitchell GF, Parise H, Benjamin EJ, et al. Changes in 7(1):99–103.
arterial stiffness and wave reflection with advancing 77. Fleg JL, O’Connor F, Gerstenblith G, et al. Impact of
age in healthy men and women: the Framingham age on the cardiovascular response to dynamic
Heart Study. Hypertension 2004;43(6):1239–45. upright exercise in healthy men and women.
63. Williams B, Lacy PS. Central haemodynamics and J Appl Physiol 1995;78(3):890–900.
clinical outcomes: going beyond brachial blood 78. Bergmann O, Bhardwaj RD, Bernard S, et al.
pressure? Eur Heart J 2010;31(15):1819–22. Evidence for cardiomyocyte renewal in humans.
64. Pearson T, Mensah G, Alexander R. Markers of Science 2009;324(5923):98–102.
inflammation and cardiovascular disease application 79. Kajstura J, Urbanek K, Perl S, et al. Cardiomyogen-
to clinical and public health practice: a statement for esis in the adult human heart. Circ Res 2010;107(2):
healthcare professionals from the Centers for 305–15.
Disease Control and Prevention and the American 80. Urbanek K, Torella D, Sheikh F, et al. Myocardial
Heart Association. Circulation 2003;107(3):499–511. regeneration by activation of multipotent cardiac
65. Najjar S, Scuteri A, Shetty V, et al. Pulse wave stem cells in ischemic heart failure. Proc Natl Acad
velocity is an independent predictor of the longitu- Sci U S A 2005;102(24):8692–7.
dinal increase in systolic blood pressure and of inci- 81. Wong LS, van der Harst P, de Boer RA, et al. Aging,
dent hypertension in the Baltimore Longitudinal telomeres and heart failure. Heart Fail Rev 2010;
Study of Aging. J Am Coll Cardiol 2008;51(14):1377. 15(5):479–86.
164 Strait & Lakatta

82. Torella D, Rota M, Nurzynska D, et al. Cardiac stem 85. Najjar S. Evolving insights into the pathophysiology
cell and myocyte aging, heart failure, and insulin-like of heart failure with preserved ejection fraction.
growth factor-1 overexpression. Circ Res 2004; Curr Cardiovasc Risk Rep 2009;3(5):374–9.
94(4):514–24. 86. Dulin BR, Haas SJ, Abraham WT, et al. Do elderly
83. Gonzalez A, Rota M, Nurzynska D, et al. Activation systolic heart failure patients benefit from beta
of cardiac progenitor cells reverses the failing heart blockers to the same extent as the non-elderly?
senescent phenotype and prolongs lifespan. Circ Meta-analysis of >12,000 patients in large-scale
Res 2008;102(5):597–606. clinical trials. Am J Cardiol 2005;95(7):896–8.
84. Juhaszova M, Rabuel C, Zorov DB, et al. Protection 87. Massie BM, Carson PE, McMurray JJ, et al. Irbesar-
in the aged heart: preventing the heart-break of old tan in patients with heart failure and preserved ejec-
age? Cardiovasc Res 2005;66(2):233–44. tion fraction. N Engl J Med 2008;359(23):2456–67.

You might also like