Download as pdf or txt
Download as pdf or txt
You are on page 1of 19

Cardiovascular Aging Compendium

Mechanisms of Vascular Aging


Zoltan Ungvari, Stefano Tarantini, Anthony J. Donato, Veronica Galvan, Anna Csiszar

Abstract: Aging of the vasculature plays a central role in morbidity and mortality of older people. To develop novel
treatments for amelioration of unsuccessful vascular aging and prevention of age-related vascular pathologies, it is
essential to understand the cellular and functional changes that occur in the vasculature during aging. In this review,
the pathophysiological roles of fundamental cellular and molecular mechanisms of aging, including oxidative
stress, mitochondrial dysfunction, impaired resistance to molecular stressors, chronic low-grade inflammation,
genomic instability, cellular senescence, epigenetic alterations, loss of protein homeostasis, deregulated nutrient
sensing, and stem cell dysfunction in the vascular system are considered in terms of their contribution to the
pathogenesis of both microvascular and macrovascular diseases associated with old age. The importance of
progeronic and antigeronic circulating factors in relation to development of vascular aging phenotypes are
discussed. Finally, future directions and opportunities to develop novel interventions to prevent/delay age-related
vascular pathologies by targeting fundamental cellular and molecular aging processes are presented.   (Circ Res.
2018;123:849-867. DOI: 10.1161/CIRCRESAHA.118.311378.)
Key Words: atherosclerosis ◼ inflammation ◼ phenotype ◼ proteostasis ◼ stem cells

C ardiovascular and cerebrovascular diseases are the most


common cause of death among older people in the United
States1 accounting for ≈1/3 of all deaths in the United States at
and pharmacological countermeasures to treat this growing
health issue.
Rapid advances in geroscience in the last 25 years, includ-
the age of 65 and ≈2/3 at an age of 85 years. With a projected ing studies on invertebrate models of aging, long-lived mam-
increase in the number of adults >65 years of age increasing mals, transgenic mouse strains, and interventional studies,
from 12% to 22% in the next 30 years, addressing age-related have led to the identification of evolutionarily conserved path-
Downloaded from http://ahajournals.org by on June 28, 2023

vascular diseases is of critical importance because the annual ways involved in life span regulation, as well as common de-
cost to care for these older people is projected to more than nominators of aging in different organisms.4 In this review, the
double over that time.2 Because many age-related cardiovas- pathophysiological roles of these aging mechanisms, including
cular and cerebrovascular diseases are because of alterations oxidative stress, mitochondrial dysfunction, impaired resis-
in arterial function or are exacerbated by arterial functional tance to molecular stressors, chronic low-grade inflammation,
and phenotypic changes, it is important to better elucidate genomic instability, telomere attrition and cellular senescence,
mechanisms underlying arterial aging.3 Furthermore, because epigenetic alterations, loss of protein homeostasis (proteosta-
the microcirculation is pervasive, being present in every tis- sis), deregulated nutrient sensing, stem cell exhaustion, and
sue in the body, it has a unique ability to influence the lo- altered intercellular communication in the vascular system, are
cal environment of the majority of tissues and organs. As considered in terms of their contribution to the pathogenesis
such, aging-induced functional and structural alterations of of both microvascular and macrovascular diseases (Figure 1).
the microcirculation contribute to the pathogenesis of range The interconnectedness between the potential mechanisms of
of age-related diseases, including vascular cognitive impair- vascular aging and the interaction between the cellular and mo-
ment, Alzheimer disease, sarcopenia, and kidney and eye dis- lecular aging processes and disease-specific pathways are dis-
ease. Therefore, it is also critical to explore the spectrum of cussed. Likewise, the implications of molecular, cellular, and
age-related microvascular functional and phenotypic changes system theories of aging for vascular aging phenotypes are con-
from subclinical dysfunction to manifested disease as to better sidered. Finally, based on our current mechanistic understand-
predict and prevent microvascular contributions to the patho- ing of vascular aging, potential novel targets for intervention to
genesis of multiple diseases associated with old age. A better improve cardiovascular and cerebrovascular health are identi-
mechanistic understanding of macrovascular and microvascu- fied. As such, it is critical to move these potential new therapies
lar aging processes is critical to find and evaluate both lifestyle forward to reduce the morbidity and mortality associated with

From the Vascular Cognitive Impairment Laboratory, Reynolds Oklahoma Center on Aging (Z.U., S.T., A.C.) and Department of Geriatric Medicine,
Translational Geroscience Laboratory (Z.U., S.T., A.C.), University of Oklahoma Health Sciences Center, Oklahoma City; Department of Medical Physics
and Informatics, University of Szeged, Hungary (Z.U., A.C.); Department of Pulmonology, Semmelweis University of Medicine, Budapest, Hungary
(Z.U.); Division of Geriatrics, Department of Internal Medicine, University of Utah, Salt Lake City (A.J.D.); Veterans Affairs Medical Center-Salt Lake
City, Geriatrics Research Education and Clinical Center, UT (A.J.D.); and Barshop Institute for Longevity and Aging Studies (V.G.) and Department of
Physiology (V.G.), University of Texas Health Science Center at San Antonio.
Correspondence to Zoltan Ungvari, MD, PhD, Department of Geriatric Medicine, Reynolds Oklahoma Center on Aging, The University of Oklahoma
Health Sciences Center, 975 NE 10th St, BRC 1303, Oklahoma City, OK 73104. Email zoltan-ungvari@ouhsc.edu
© 2018 American Heart Association, Inc.
Circulation Research is available at https://www.ahajournals.org/journal/res DOI: 10.1161/CIRCRESAHA.118.311378

849
850  Circulation Research  September 14, 2018

by increased oxidative stress contributes significantly to both


Nonstandard Abbreviations and Acronyms
impaired dilation of coronary arteries,13 promoting myocar-
ACE angiotensin-converting enzyme dial ischemia, and neurovascular uncoupling, impairing the
AMPK AMP-activated protein kinase moment-to-moment adjustment of cerebral blood flow to in-
ApoE apolipoprotein E creased oxygen and nutrient demand that occurs with neuro-
eNOS endothelial NO synthase
nal activation.31,32 In addition to inactivation of NO by ROS,
EPC endothelial progenitor cell
alterations in eNOS (endothelial NO synthase) activation
status, substrate (L-arginine) and cofactor (BH4) availability,
IL interleukin
increased endothelin-1,26 and reduced expression of eNOS33–35
iNOS inducible NO synthase
may also contribute to age-related reduction in NO bioavail-
miRNA microRNA
ability. NO exerts potent anti-inflammatory, antithrombotic,
MLKL mixed-lineage kinase domain like
and antileukocyte adhesion effects; thus reduction in NO like-
MMP matrix metalloproteinase
ly promotes a proatherogenic vascular phenotype in aging.36–38
mtDNA mitochondrial DNA
Many of the results of vascular oxidative stress are mediated
mTOR mechanistic/mammalian target of rapamycin via production of the highly reactive oxidant peroxynitrite—the
mtROS mitochondrial reactive oxygen species reaction product of NO and superoxide,39,40 which has been well
RAS renin-angiotensin system documented in older endothelial cells and arteries.13,15,20,22 The
ROS reactive oxygen species mechanisms by which peroxynitrite contributes to vascular ag-
TAME Targeting Aging With Metformin ing are multifaceted and include direct cytotoxic effects, adverse
TGF-β transforming growth factor-β effects on mitochondrial function, and activation of inflamma-
TLR toll-like receptor tory pathways (Figure 2). In particular, oxidative stress and the
TNF-α tumor necrosis factor-α consequent activation of redox-sensitive cellular signaling path-
VSMC vascular smooth muscle cell ways, including NF-κB (nuclear factor kappa-light-chain-en-
hancer of activated B cells), are thought to be implicated in the
cardiovascular and cerebrovascular dysfunction/disease and inflammatory process in the aged vasculature. This inflammato-
thereby improving health span in an increasingly aged popu- ry process is characterized by increased endothelial activation41
lation. We will not discuss in detail the data on the effects of and proinflammatory changes in the cytokine expression profile
preventive measures already available and in clinical use (in- of aged vascular cells.18,19,42 Increased vascular oxidative stress
cluding physical exercise, smoking cessation, dietary regimens, has also been linked to activation of MMP (matrix metallopro-
Downloaded from http://ahajournals.org by on June 28, 2023

inhibitors that disrupt the renin-angiotensin system [RAS], and teinases) and consequential disruption of the structural integrity
statins) on vascular aging phenotypes, as these have been the of aged arteries, potentially contributing to large artery stiffen-
subject of a recent comprehensive reviews.5–8 The great deal ing43 and the pathogenesis of aortic aneuryms.44 Furthermore,
of phenomenological work has been performed on the effects in the cerebral circulation, aging- and hypertension-dependent
of aging on specific cell types within the vascular wall and on activation of the ROS-MMP axis promotes the development
structural alterations and the hemodynamic consequences that of cerebral microhemorrhages,45 which contribute to cognitive
result from arterial stiffening. We refer the interested reader to decline, geriatric psychiatric syndromes, and gait disorders.46
excellent reviews on these topics.6,9–11 Importantly, in preclinical models, similar antioxidative treat-
ments were reported to prevent large artery stiffening,47 cerebral
Molecular and Cellular Mechanisms microhemorrhages,45 and aortic aneurysms,44 providing further
of Vascular Aging evidence for shared pathomechanisms.

Role of Oxidative and Nitrative Stress Role of Mitochondrial Dysfunction


Since Denham Harman first proposed the free radical theory Mitochondria play a central role in regulation of aging pro-
of aging in the 1950s,12 a large amount of data has been pub- cesses,4,48 including regulation of life span.49 As mammals age,
lished implicating oxidative stress in vascular aging processes the efficacy of the respiratory chain diminishes, promoting
(Figure 2). In particular, there is strong evidence that increased electron leakage and increased ROS production and reducing
production of reactive oxygen species (ROS) by NADPH cellular ATP generation.
(reduced nicotinamide adenine dinucleotide phosphate) oxi- Recent studies suggest that mitochondrial ROS (mtROS)
dases,13–17 and mitochondria18,19 contributes to endothelial dys- production has an important role in age-related vascular dys-
function and large elastic artery stiffening with advancing age function.18,24,50,51 In the aged vasculature, increased mtROS has
both in laboratory animals13,14,20–24 and humans.16,25 Although been attributed to a dysfunctional electron transport chain52
oxidative stress may influence many facets of vascular func- and is likely exacerbated by peroxynitrite-mediated nitra-
tion with advancing age via oxidation of critical proteins or tion and inhibition of MnSOD (manganese-dependent super-
induction of redox-sensitive transcription factors, one of its oxide dismutase),14 decline in cellular glutathione content,42
most potent effects is inactivation of endothelium-derived NO. downregulation of p66Shc,53 or impaired Nrf2 (nuclear factor
Impaired bioavailability of NO is responsible for age-related [erythroid-derived 2]-like 2)-mediated antioxidant defense re-
reduction in endothelium-dependent dilation, enhanced vaso- sponses.18,54,55 There is also evidence that aging is associated with
constriction, and dysregulation of tissue perfusion.13,14,20,26–30 impaired mitochondrial biogenesis in endothelial and smooth
There is strong evidence that endothelial dysfunction caused muscle cells both in conduit arteries52 and the capillaries,56,57
Ungvari et al   Mechanisms of Vascular Aging   851
Downloaded from http://ahajournals.org by on June 28, 2023

Figure 1. Conceptual model for the role of cell-autonomous and noncell-autonomous mechanisms in vascular aging. The model predicts that circulating
progeronic (eg, inflammatory cytokines, renin-angiotensin system [RAS], and aldosterone) and antigeronic factors (eg, IGF-1 [insulin-like growth factor 1],
mediators of caloric restriction, estrogen) derived from the brain, the endocrine system, cells of the immune system, and the adipose tissue orchestrate aging
processes simultaneously in the endothelial and smooth muscle cells within the large vessels and microcirculation. The hierarchical regulatory cascade for
vascular aging involves modulation of cell-autonomous cellular and molecular aging processes. The resulting functional dysregulation of vascular cells (ie,
impaired vasomotor, barrier, secretory, and transport functions of the vasculature, as well as adverse structural remodeling) promotes the development of a wide
range of age-related vascular pathologies. AMI indicates acute myocardial infarction; GDF11, growth differentiation factor 11; and PAD, peripheral artery disease.

which is likely to negatively impact cellular energetics and also to restore endothelium-dependent regulation of cerebral blood
may increase mtROS production by increasing electron flow flow via neurovascular coupling, improving cognitive function
through the deficient electron transport chain. Importantly, in aged mice.51 There is evidence that mitochondria-derived
mtROS can be pharmacologically targeted for vasoprotection. H2O2 promotes low-grade vascular inflammation in aging by
For example, treatment with the mitochondrial antioxidant inducing NF-κB activation in endothelial and smooth muscle
MitoQ,58 resveratrol59 (which substantially attenuates mtROS cells19,24 (Figure 2). Recent studies also link increased hyper-
production in endothelial and smooth muscle cells19,60), and the tension-induced mtROS production in aged vascular smooth
potent mitochondria-targeted antioxidative tetrapeptide SS-3151 muscle cells (VSMCs)50 to increased MMP activation in the
has been shown to improve endothelial function in arteries from vascular wall and consequential exacerbation of cerebral mi-
rodent models of aging. Treatment with SS-31 was also shown crohemorrhages.46 Another potentially important link between
852  Circulation Research  September 14, 2018

Figure 2. Proposed scheme for mechanisms and pathological consequences of age-related oxidative stress in vascular endothelial cells. The
model predicts that in aged endothelial cells, dysfunctional mitochondria and NADPH (reduced nicotinamide adenine dinucleotide phosphate) oxidases are
critical sources of increased reactive oxygen species (ROS) production. Increased levels of O2.− generated by the electron transport chain are dismutated
to H2O2, which can penetrate the mitochondrial membrane increasing cytoplasmic H2O2 levels. Increased oxidative stress is exacerbated by age-related
impairment of Nrf2 (nuclear factor [erythroid-derived 2]-like 2)-dependent homeostatic antioxidant defense mechanisms. H2O2 plays important signaling
roles, including activation of NF-κB (nuclear factor kappa-light-chain-enhancer of activated B cells), which contribute to age-associated low-grade chronic
vascular inflammation. Increased levels of O2.− generated by NADPH oxidases (stimulated by elevated TNF-α [tumor necrosis factor-α] levels or by the
activated local renin-angiotensin system [RAS] in the vascular wall) decrease the bioavailability of NO by forming peroxynitrite (ONOO−). Increased nitrative
stress leads to PARP-1 activation, which promotes vascular inflammation and contributes to cellular energetic dysfunction by consuming NAD+ (nicotinamide
adenine dinucleotide), compromising sirtuin-mediated antiaging pathways. Impaired bioavailability of NO promotes vasodilator dysfunction and compromises
endothelial viability. In addition, increased vascular oxidative stress in aging also induces MMP (matrix metalloproteinase) activation, promoting the
Downloaded from http://ahajournals.org by on June 28, 2023

pathogenesis of intracerebral hemorrhages, aneurysm formation, and blood-brain barrier disruption. CMV indicates cytomegalovirus; PARP-1, poly (ADP-
ribose) polymerase 1; SASP, senescence-associated secretory phenotype; SIRT1, sirtuin1; and TLR, toll-like receptor.

mtROS production and vascular aging is the induction of apop- mtDNA polymerase (polG [DNA polymerase subunit gamma])
tosis via a Bcl-2 (B-cell lymphoma 2)–dependent pathway.61 deficient in proof-reading activity accumulate mutations in their
It is increasingly realized that the functional integrity of the mtDNA and exhibit accelerated atherosclerosis, associated with
vasculature, including regulation of membrane transport and impaired proliferation and apoptosis of VSMCs.65
barrier functions, depends of normal cellular energy metabolism. The NAD+ (nicotinamide adenine dinucleotide)-dependent
Thus, dysfunctional mitochondria and impaired mitochondrial prosurvival enzyme SIRT1 (sirtuin1) modulates mitochondrial
energy metabolism can potentially contribute to vascular aging function in the vasculature, controlling mitochondrial biogen-
in addition to the effects of increased mitochondrial release of esis, mtROS production, and cellular energy metabolism,42,60,66
ROS. The mitochondrial DNA (mtDNA) has a high mutation as well as removal of damaged mitochondria by autophagy.67
rate, because of the proximity of mtDNA to sites of ROS pro- The mitochondrial sirtuin SIRT3 also regulates many key en-
duction in the mitochondria, the lack of protective histone cover- zymes involved in mitochondrial energy metabolism. NAD+
age in the mtDNA, and the limited efficiency of mtDNA repair is a rate-limiting cosubstrate for sirtuin enzymes, and there is
mechanisms. There is growing evidence that aging increases growing evidence that cellular NAD+ availability decreases in
mutations and deletions in mtDNA, eroding mitochondrial en- aging,68,69 at least in part, because of overactivation of NAD+
ergy production and contributing to aging processes.48 mtDNA utilizing PARP-1 (poly [ADP-ribose] polymerase 1).41 There
damage is likely a result of increased mitochondrial oxidative is strong evidence that in aged mice, enhancing NAD+ biosyn-
stress.62 Although aging is associated with significant mitochon- thesis (eg, by treatment with nicotinamide mononucleotide—a
drial oxidative stress in the vasculature,18,19,24,50 the role of mtD- key NAD+ intermediate68) rescues age-related functional altera-
NA damage in the development of vascular aging phenotypes tions in the aorta70 and cerebral vasculature (Tarantini, Csiszar,
is not well understood. To date, only few studies have focused and Ungvari; unpublished findings, 2017), likely by activating
on the link between mitochondrial heteroplasmy and atherogen- sirtuin-mediated pathways and reversing age-related decline
esis.63 mtDNA deletions have been detected in human athero- in mitochondrial function.71 Other potential mechanisms con-
sclerotic lesions,63,64 which may contribute to impaired cellular tributing to impaired bioenergetics in aged cells include oxida-
metabolism in the vascular wall. Recent studies provide direct tion/nitration of mitochondrial proteins, destabilization of the
evidence that mitochondrial mutations play a causal role in ath- macromolecular organization of electron transport chain com-
erogenesis in mouse models of the disease.65 Importantly, trans- plexes, and impaired mitophagy (a mitochondria-specific form
genic mice ApoE−/− (apolipoprotein E) harboring a version of the of autophagy). The combination of increased mitochondrial
Ungvari et al   Mechanisms of Vascular Aging   853

damage and decreased turnover of mitochondria, because of inflammatory processes in the aged vascular wall.41 First, ROS
impaired biogenesis and deficient mitophagy, likely contrib- act as signaling molecules activating proinflammatory signaling
utes to the accumulation of dysfunctional mitochondria in the pathways, including NF-κB,24 which regulate endothelial acti-
vascular cells, exacerbating vascular aging processes. vation and expression of proinflammatory paracrine mediators
and promote atherogenesis. Importantly, aged endothelial and
Vascular Inflammation in Aging
smooth muscle cells exhibit significant NF-κB activation,24,54
There is strong experimental and clinical evidence that chronic,
and selective inhibition of NF-κB in the vasculature was shown
sterile, low-grade inflammation is a hallmark of the aging pro-
to improve blood flow regulation, decrease systemic inflamma-
cess (inflammaging72). Age-related activation of inflammatory
tion, exert beneficial metabolic effects, and extend health span.78
processes plays a key role in a wide range of macrovascular and
Second, inflammatory mediators are potent inducers of cellular
microvascular pathologies, ranging from atherogenesis and an-
eurysm formation to microvascular dysfunction, blood-brain oxidative stress (eg, TNF-α activates NADPH oxidases35).
barrier disruption, and Alzheimer pathologies41 (Figure 3). SIRT1 exerts potent anti-inflammatory effects, and de-
Previous studies demonstrate that both in aged laboratory ro- creased SIRT1 activity likely contributes to vascular inflam-
dents and primates, there is a proinflammatory shift in the gene mation in aging.42,79,80 Importantly, pharmacological activators
expression profile of vascular endothelial and smooth muscle of SIRT1 were shown to attenuate vascular inflammation in
cells, including an induction of inflammatory cytokines (eg, IL aged mice.59,80
[interleukin]-6, IL-1β, and TNF-α [tumor necrosis factor-α]), Endothelial senescence (see below) is associated by
chemokines, adhesion molecules, iNOS (inducible NO syn- a significant increase in the production/release of a wide
thase), and other proinflammatory mediators.13,24,33,59,73–76 The range of inflammatory cytokines and chemokines,81 termed
resulting proinflammatory microenvironment in the vascular the senescence-associated secretory phenotype. Induction
wall promotes vascular dysfunction,35,77 impairs cellular me- of senescence-associated secretory phenotype is likely me-
tabolism, increases apoptosis,35,74 and contributes to the patho- diated by activation of NF-κB, p38MAPK, the DNA dam-
genesis of vascular diseases. age response pathway, and GATA4 (transcription factor
The mechanisms contributing to vascular inflammation GATA-4).82,83 There is growing evidence that the presence
in aging are likely multifaceted. There is an important cross- of senescent cells contributes significantly to the heightened
talk between increased oxidative stress and activation of inflammatory status of the aged vasculature.84
Downloaded from http://ahajournals.org by on June 28, 2023

Figure 3. Mechanisms and consequences of age-related vascular inflammation. The model predicts that multiple pathways converge on activation of
inflammatory processes in the vascular tissue. During aging, increased reactive oxygen species (ROS) production, exacerbated by Nrf2 (nuclear factor [erythroid-
derived 2]-like 2) dysfunction, enhances NF-κB (nuclear factor kappa-light-chain-enhancer of activated B cells) activation, which promotes inflammatory cytokine
and chemokine expression, microvascular endothelial activation, leukocyte adhesion, and extravasation. Increased nitrative stress promotes PARP-1 activation,
which contributes to impaired activity of anti-inflammatory sirtuins. Sterile inflammation in the vascular wall is also exacerbated by increased secretion of
inflammatory mediators from senescent cells and danger-associated molecular patterns, which activate innate immune system effectors, including TLRs (toll-
like receptors) and the NLRP3 (NACHT, LRR, and PYD domains-containing protein 3) inflammasome complex. The aging vasculature in humans is also affected
by the high prevalence of endothelium-trophic persistent cytomegalovirus infection. Inflammatory processes contribute to a wide range of macrovascular and
microvascular pathologies affecting older people. eNOS indicates endothelial NO synthase; GSH, glutathione; IkBa, nuclear factor of kappa light polypeptide
gene enhancer in B-cells inhibitor, alpha; iNOS, inducible NO synthase; L-arg, L-arginine; MMP, matrix metalloproteinase; mtDNA, mitochondrial DNA; mtROS,
mitochondrial ROS; NAD, nicotinamide adenine dinucleotide; Nrf2, nuclear factor (erythroid-derived 2)-like 2; ONOO, peroxynitrite; PARP-1, poly (ADP-ribose)
polymerase 1; RAS, renin-angiotensin system; SIRT1, sirtuin1; TNF-α, tumor necrosis factor-α; and TNFR, tumor necrosis factor receptor.
854  Circulation Research  September 14, 2018

Sterile inflammation in the vascular wall is also exacerbated anti-inflammatory94 and proangiogenic95 effects. There is
by danger-associated molecular patterns, which activate innate strong evidence that aging promotes Nrf2 dysfunction in
immune system effectors, including TLRs (toll-like receptors) the vasculature, exacerbating oxidative stress and increas-
and the NLRP3 (NACHT, LRR, and PYD domains-containing ing sensitivity of aged vascular cells to ROS-mediated cel-
protein 3) inflammasome complex. TLRs are activated by a lular and molecular damage.54,55 This loss of oxidative stress
wide range of danger-associated molecular pattern ligands (eg, resilience may be a major determinant for the development
molecules released to the extracellular matrix from necrotic of age-related vascular pathologies.96 Importantly, antiaging
cells, breakdown products of the extracellular matrix, or bacte- effects of caloric restriction is associated with induction of
rial breakdown products leaking through a damaged gut barrier) Nrf2-mediated pathways.18,97 Further studies are warranted
and control the secretion of several proinflammatory paracrine to determine how pharmacological activation of Nrf2 exerts
mediators (eg, IL-1, IL-6, IL-8, and TNF-α). There is evidence antiaging vasoprotective effects.
that aging induces a proinflammatory phenotype in VSMCs, at
least in part, because of activation of TLR4-mediated, MyD88- Loss of Proteostasis in Vascular Aging
dependent signaling pathways.76 Strong data suggest that the Impaired maintenance of proteostasis is thought to contribute
canonical Nlrp3 inflammasome contributes to systemic low- to organismal aging.98 There is evidence that activity of pro-
grade age-related sterile inflammation in mice,85 but further teostasis networks and proteome stability determine healthy
studies are needed to establish the exact role of this mechanism cardiac aging.99,100 It is suspected that disequilibrium between
in age-related vascular pathologies. protein synthesis, maintenance, and degradation also compro-
There is growing evidence that impaired oxidative stress mises vascular health. Indeed, increased presence of misfolded
resilience in aging also exacerbates vascular inflammation in- protein aggregates is associated with cardiovascular diseases.101
duced by cardiovascular risk factors, including obesity, meta- Aging impacts multiple components of the proteostasis sys-
bolic disease, and hypertension.86–88 The interaction between tems in heart and the vasculature, including chaperones,102 the
aging and the effect of environmental inflammatory factors ubiquitin-proteasome, and the lysosome-autophagy system.98
(eg, particulate exposure89) to exacerbate vascular inflamma- Chaperones assist the folding, assembly, disassembly, and
tion warrants further studies. transport of other proteins and play a major role in preventing
Further studies are needed to differentiate between adaptive protein misfolding and aggregation. Many age-related molecu-
and maladaptive inflammatory responses in the aged vascula- lar alterations can impact chaperoning activities. For example,
ture and to define the role of chronic low-grade microvascu- aging results in downregulation of HSP70 (70 kilodalton heat
lar inflammation in a wide range of functional impairments shock proteins) in the vascular tissue.102 Further, mitochondrial
Downloaded from http://ahajournals.org by on June 28, 2023

in multiple organs. The interaction between cell-autonomous dysfunction and the resulting decline in cellular ATP content
mechanism of age-related vascular inflammation and inflam- likely also impair the function of ATP-dependent chaperones.
matory processes induced by bacterial breakdown products The processes of autophagy (macroautophagy, microautopha-
getting to the circulation through a leaky intestinal barrier or gy, and chaperone-mediated autophagy) allow the degradation
by chronic infection with viruses that exhibit endothelial tro- and recycling of cellular constituents. Recently, hypotheses
pism in vascular inflammation also has to be further clarified. were put forward that dysregulation of autophagic processes,
For example, >90% of adults ≥80 years of age have persistent including mitophagy, may be a common pathway promoting
human cytomegalovirus infection.90 Cytomegalovirus repli- vascular aging and development of age-related vascular dis-
cates in the vascular endothelial cells during the entire life of eases.103 Experimental findings showing increased oxidative
the host after initial infection, and severity of cytomegalovirus stress, impaired bioavailability of NO, and upregulation of in-
infection (antibody titers) was shown to predict increased in- flammatory mediators in autophagy-deficient endothelial cells
cidence of frailty and risk of mortality.91 support this view.104 Further, pharmacological interventions
that stimulate autophagy (eg, trehalose or spermidine treat-
Maladaptation to Molecular Stresses: Role of Nrf2 ment) were reported to reverse aspects of arterial aging.105,106
Dysfunction Proteasomes degrade unneeded or damaged proteins by pro-
Recent progress in geroscience research has identified a teolysis. There is evidence that proteasome activity declines in
critical hallmark of the aging process: impaired ability of advanced aging98 and is diminished in atherosclerotic plaques
aged cells to respond to molecular stresses and return to of elderly patients107 and in hearts of aging rats.108 In addition to
homeostasis. In young organisms, in response to increased direct effects on protein degradation, the ubiquitin-proteasome
generation of ROS in the vascular endothelial and smooth system is critical for the activation of key regulators of ath-
muscle cells, adaptive homeostatic mechanisms are invoked erogenesis and vascular inflammation.109 Future studies should
that involve activation of Nrf2-driven antioxidant defense identify the role of the ubiquitin-proteasome system and other
pathways.54,55,92 Nrf2 is an evolutionarily conserved redox- proteostatic pathways that are impaired in specific vascular
sensitive transcription factor, which coordinates the anti- disease states associated with aging and elucidate age-related
oxidant response, including upregulation of enzymes that factors, including neuroendocrine factors, which regulate pro-
detoxify ROS and repair ROS-induced macromolecular teostasis machineries in the vascular wall.
damage.93 In the young vasculature, this adaptive homeostat-
ic response serves to reduce oxidative stress and attenuate Role of Genomic Instability
cellular and macromolecular damage caused by increased Since the original formulation of Somatic Mutation Theory
ROS levels. Induction of Nrf2 was also shown to exert potent of aging,110 a large amount of data has been published both in
Ungvari et al   Mechanisms of Vascular Aging   855

support and against a causal role of DNA damage and muta- phenotypic changes in VSMCs121,122; however, it remains to be
tion accumulation in aging.111 There is evidence that diverse demonstrated that these pathways also contribute to normal
genetic lesions may accumulate within aged cells, including vascular aging.
somatic mutations, chromosomal aneuploidies, and copy Impairment of mechanisms responsible for maintain-
number variations and telomere shortening. Despite these ing the appropriate length and functionality of telomeres is
advances, the role of genomic instability in vascular aging thought to play a role in vascular aging and hypertension by
is not well understood. Hypotheses that predict that genomic inducing cellular senescence (see below).84,123
instability play a role in vascular aging, usually focus on the
primary role of oxidative stress-induced DNA damage, illus- Role of Cellular Senescence
trating how these hypotheses and the oxidative stress hypoth- Cellular senescence is a fundamental aging process in which
esis of aging are interconnected.112 Importantly, endothelial cells, including vascular endothelial and smooth muscle cells,
cells seem to have less efficient DNA repair pathways than permanently withdraw from the cell cycle in response to a
many other cell types.81 In that regard, it is interesting that range of endogenous and exogenous stressors (eg, ROS, dys-
known interventions that cause extensive DNA damage (eg, functional telomeres, DNA damage, and paracrine signals)
whole-brain irradiation) result in significant phenotypic and and undergo distinctive phenotypic alterations, including
functional alterations in endothelial cells, promoting micro- profound proinflammatory secretome changes124 (Figure 4).
vascular rarefaction, impaired vasodilation, and proinflam- Recent studies demonstrate that elimination of senescent
matory changes, mimicking several aspects of the aging cells expressing p16INK4A extends life span and health span
phenotype.81,113,114 In vascular endothelial cells, DNA damage in mice,125,126 suggesting that cellular senescence plays a fun-
readily triggers replicative senescence (see below),81 to pre- damental role in physiological decline associated with aging.
vent propagation of damaged DNA. In light of recent devel- The available evidence indicates that endothelial senescence
opments, it seems to be likely that induction of senescence also contributes to endothelial dysfunction in aging and
is a major mechanism by which DNA damage contributes to pathophysiological conditions associated with accelerated
vascular aging.115 vascular aging.114,127–129 Endothelial senescence has also been
To elucidate the causal role of DNA damage in vascular implicated in the pathogenesis of heart failure.130 Replicative
aging, an interesting recent study reported that mouse models senescence may also be potentially important for impairment
with genomic instability resulting from the defective nucleo- of regenerative and angiogenic capacity of the vascular endo-
tide excision repair genes (ERCC1 and XPD) exhibit aging- thelium. Studies using mouse models of irradiation-induced,
like vascular phenotypes, including endothelial dysfunction, DNA damage-mediated senescence81 demonstrate that induc-
Downloaded from http://ahajournals.org by on June 28, 2023

increased vascular stiffness, increased presence of senescence tion of cellular senescence in the neurovascular unit associ-
cells, and hypertension.116 However, these mouse models also ates with significant cerebromicrovascular dysfunction114 and
exhibit severe liver, kidney, bone marrow, neurological, or microvascular rarefaction, mimicking the aging phenotype.
bone phenotypes, which associate with reduced life span,116 Advanced atherosclerotic lesions contain senescent cells,
and it is unclear how closely these phenotypes indeed mimic and recent studies using genetic and pharmacological ap-
aging. Mice with genetic deficiency in the spindle assembly proaches to eliminate senescent cells in Ldlr−/−mice suggest
checkpoint protein BubR1 (budding uninhibited by benzimid- that senescent cells promote atherogenesis, contributing to
azole-related 1), which promotes progressive aneuploidy, were plaque instability by upregulating MMPs131 or by exacerbat-
also shown to exhibit aging-like vascular phenotypes, includ- ing vascular inflammation.132 The hypothesis was put for-
ing endothelial dysfunction, increased vascular stiffness, me- ward that pharmacological treatment with senolytic agents to
dia atrophy, and fibrosis.117 However, this mouse model also clear senescence cells may exert atheroprotective effects.131
exhibits short life span associated with severe functional defi- Importantly, long-term senolytic treatment was also shown
cits in multiple organs, including cachectic dwarfism and lor- to improve endothelial function in mouse models of aging.127
dokyphosis. Thus, the relevance of this model to normal aging However, the exact role of different senescence mechanisms
remains unresolved. It was also proposed that an association in age-related vascular pathophysiology is not well under-
exists between single-nucleotide polymorphisms in human stood. Future studies should address the relationship among
DNA repair genes and vascular stiffness,116 yet the mechanis- acquisition of a senescence-associated secretory phenotype
tic role of DNA repair pathways in the genesis of age-related in the endothelium and the VSMCs and specific disease pro-
vascular diseases in humans remains to be determined. There cesses. There is evidence that the senescence-associated se-
are studies reporting that increased oxidative DNA damage118 cretory phenotype can also induce paracrine senescence and
and increased expression of multiple biomarkers of DNA alter the function of neighboring cells, and the role of this
double-strand breaks119 are present in atherosclerotic plaques. mechanism in vascular aging should be further evaluated.
Although accelerating or retarding repair of double-strand The possibility of paracrine transmission of senescence from
breaks in transgenic mouse models have lesser effect on ath- microvascular endothelial cells to parenchymal cells also
erogenesis, they significantly alter plaque stability.119 requires further investigations. It should be noted that many
Children with Hutchinson-Gilford progeria syndrome and studies assess only senescence-associated β-galactosidase ac-
other laminopathies exhibit accelerated vascular pathologies tivity as a marker of senescent cells. Future studies assessing
leading to fatal myocardial infarction or stroke at a young should use novel molecular markers of senescence and senes-
age.120 There is evidence implicating DNA damage response cence reporter mouse models and analyze senescence-related
induced by genetic nuclear lamina dysfunction in aging-like gene expression in individual cells.
856  Circulation Research  September 14, 2018

Figure 4. Conceptual model for the pathogenic role of cellular senescence in vascular aging. The model predicts that increased presence of
senescent endothelial or smooth muscle cells (SMCs) in the aged vasculature and their proinflammatory secretome (SASP [senescence-associated secretory
phenotype]) contributes to impaired angiogenesis and microvascular rarefaction, pathological remodeling of the extracellular matrix (ECM), barrier disruption,
chronic inflammation, and atherogenesis. MMP indicates matrix metalloproteinase.

Role of Increased Apoptosis and Necroptosis upregulation of necroptosis pathways promotes aorta aneu-
Apoptosis is an evolutionarily conserved cell death program rysm progression in mouse models.137
that is tightly regulated and executed through the interac- Taken together, pathways involved in programmed cell
Downloaded from http://ahajournals.org by on June 28, 2023

tion of extrinsic and intrinsic signaling pathways.133 There death are promising targets for interventions in multiple ag-
is strong evidence that alterations in apoptotic potential con- ing-related diseases, including cardiovascular protection, as
tribute to several aging phenotypes across species, including well as prevention of cell loss leading to muscle atrophy and
the genesis of age-related cardiovascular pathologies. In the neurodegeneration.
vasculature, there is an increased presence of apoptotic endo-
thelial cells, which has been attributed to impaired bioavail- Role of Epigenetic Alterations
ability of NO, upregulation of TNF-α, and mitochondrial A wide range of epigenetic alterations affects the cells dur-
oxidative stress.35,59,74,134 Increased apoptotic cell death likely ing the life span, which may modulate vascular aging pheno-
contributes to aging-induced microvascular rarefaction and types.138 Epigenetic changes that may contribute to vascular
the pathogenesis of atherosclerotic vascular diseases and an- aging processes involve alterations in DNA methylation pat-
eurysm formation.11 terns, posttranslational modification of histones, microRNAs
Necroptosis is a newly identified form of programmed (miRNAs), long noncoding RNAs, and chromatin remodeling.
cell death that does not involve caspase activation but criti- DNA methylation is thought to be a central regulator of
cally depends on receptor-interacting serine-threonine ki- genome function. Aging is associated with complex, likely
nase 3 and MLKL (mixed-lineage kinase domain like) and sexually dimorphic alterations in methylation patterns in
is characterized by morphological features of necrosis.135 many organs, which can be partially reversed by antiaging
Necroptosis plays a role in inflammaging by promoting interventions (eg, caloric restriction).139 Also, in animal mod-
proinflammatory phenotypic changes in tissues because of els of aging, altered methylation of genes important for vas-
the release of cell debris (danger‐associated molecular pat- cular function has been observed.140,141 There is preliminary
terns).136 Danger-associated molecular patterns are a major evidence that in vascular diseases, DNA methylation patterns
activator of NLRP3 inflammasome, which is an important of cells within the vascular wall are altered142; however, un-
mechanism involved in low‐grade chronic inflammation in derstanding the pathogenic role of these changes in vascular
aging (see above). Inhibition of necroptosis either geneti- aging requires further studies.
cally, pharmacologically, or by dietary means was shown Understanding how posttranslational histone modifica-
to reduce inflammation in mouse models.135,136 Importantly, tions (lysine methylation and acetylation), which were dem-
biomarkers of necroptosis are evident in the atherosclerotic onstrated to regulate life span in lower organisms and to
plaques in ApoE-knockout mice, and inhibition of necrop- modulate aging phenotypes in mammals, modulate vascular
tosis pathways reduces atherosclerosis burden and increases aging processes is an area of intense current research.4 Histone
the life span in these models.135 Biomarkers of necroptosis acetylation is dynamically regulated by histone acetyltransfer-
are also increased in human aorta aneurysmal tissues, and ases and histone deacetylases. There is particularly strong data
Ungvari et al   Mechanisms of Vascular Aging   857

that decreased activity/expression of class III histone deacety- by controlling cellular responses to nutrient availability and
lases (the NAD+ utilizing sirtuin family) contributes to vascu- growth signals, including mTOR (mechanistic/mammalian
lar aging.42,70,79,80,143 target of rapamycin) signaling,153 AMPK (AMP-activated pro-
DNA- and histone-modifying enzymes act in concert with tein kinase), and sirtuins.
key epigenetic factors that determine changes in chromatin ar- It is now recognized that mTOR acts as a critical molecu-
chitecture, regulating life span and health span in evolution- lar regulator of key anabolic processes, controlling biosyn-
arily diverse organisms.4 Such chromatin remodeling factors thesis of proteins, lipids, and nucleic acids. In low nutrient
include the Polycomb group proteins, which can remodel chro- conditions, mTOR activity is reduced, which relieves mTOR-
matin such that epigenetic silencing of genes takes place. The dependent inhibition of autophagy—a major catabolic cellu-
role of the Polycomb group proteins in regulation of endothelial lar process. Reduced activity of the mTOR pathway has been
progenitor cell (EPC) function has been extensively studied.144 reproducibly shown to regulate aging and extend life span in
There is evidence that expression/activity of many of these invertebrates and in mice.154 Consistent with the concept that
chromatin remodeling factors are altered in aging,4 yet, their mTOR regulates fundamental molecular processes of aging,
mechanistic role in vascular aging has yet to be elucidated. there is growing evidence that attenuating mTOR activity in-
The expression of 60% of human protein coding genes hibits or delays the pathogenesis of age-associated diseases,
is controlled through posttranscriptional repression by miR- including Alzheimer disease.155,156 There is growing evidence
NAs—a class of small noncoding RNAs. There is a complex that mTOR inhibition also confers protective, antiaging vas-
interplay between miRNAs and other epigenetic factors, and cular effects, delaying endothelial cell senescence157,158 and
dysregulation of miRNA expression is an emerging field in promoting endothelium-mediated, NO-dependent vasodi-
age-related epigenetics. In the vasculature, miRNAs con- lation.156,159–162 mTOR inhibition also regulates phenotypic
tribute to the regulation of important biological processes, switch of VSMCs.163 Recently, it was reported that chronic
including angiogenesis,145 atherogenesis,146 and restenosis.147 treatment with the mTOR inhibitor rapamycin reverses age-
There is growing evidence that aging is associated with dys- associated arterial dysfunction, decreasing vascular stiffness
regulation of miRNA expression in vascular endothelial and and oxidative stress.164 The potential significance of these
smooth muscle cells, which likely contributes to age-related findings for human aging stems from the availability of in-
impairment of angiogenic processes,148 decreased cellular hibitors for mTOR signaling pathways that are approved for
stress resilience,18 and plaque formation, destabilization, and clinical use. There is considerable excitement about the po-
rupture.149 Importantly, there is also evidence that IGF-1 (in- tential of mTOR inhibitors to treat cancer and neurological
sulin-like growth factor 1) deficiency during a critical period diseases of aging and potentially to improve health span in
Downloaded from http://ahajournals.org by on June 28, 2023

early in life results in persistent changes in posttranscriptional elderly patients. Preliminary preclinical studies suggest that
miRNA-mediated control of critical target genes for vascular mTOR inhibition may also exert certain beneficial effects on
health, which likely contribute to the deleterious late-life car- cardiovascular pathologies associated with old age, including
diovascular effects known to occur with developmental IGF-1 stroke.165–168 More importantly, recent studies156,169 identified
deficiency.150 mTOR as a major regulator of brain vascular damage and dys-
Long noncoding RNAs are transcripts >200 nucleotides, function in Alzheimer disease models156,170–172 and in models
which regulate multiple aspects of RNA transcription and of atherosclerosis.173 In mouse models of Alzheimer disease,
translation. There is growing evidence that long noncoding mTOR inhibition with rapamycin was shown to reduce Aß
RNAs interact with proinflammatory signaling pathways and vascular pathology and improve cerebral blood flow via an
regulate senescence; however, their role on regulation of vas- endothelial NO-dependent mechanism,156,169 significantly im-
cular aging processes is virtually unknown.151 Interestingly, proving cognitive outcomes. Thus, although further studies
there is initial evidence linking the expression of the long are needed to define the role of mTOR in vascular aging, the
noncoding RNA Meg3 (maternally expressed 3) to age-related available evidence indicates that mTOR has an important role
impairment of angiogenic capacity of endothelial cells.152 in cerebrovascular dysfunction associated with neurodegen-
Further studies are definitely needed to understand the erative diseases.
contribution of alteration of epigenetic patterns to the devel- Sirtuins and AMPK are critical cellular energy sensors
opment of various age-related vascular pathologies and to that are activated by cellular metabolic factors, such as NAD+
elucidate age-related changes in cellular mechanisms assur- in the case of sirtuins and the AMP:ATP ratio for AMPK.
ing the generation and maintenance of epigenetic patterns (eg, These pathways are thought to be activated in times of low
DNA methyltransferases, histone acetylases and deacetylases, nutrient status to provide the cell with enhanced stress resis-
methylases, and demethylases, protein complexes implicated tance to prevent cellular loss or derangement. Both pathways
in chromatin remodeling). Epigenetic alterations are revers- been shown to be enhanced in response to longevity pro-
ible, thus efforts should persist to develop epigenome-influ- moting interventions (eg, calorie restriction) and are known
encing interventions for prevention/treatment of age-related to interact with one another. Activators of both the sirtuins,
vascular diseases. specifically SIRT1 and SIRT6, and AMPK have been ef-
fective in improving endothelial function, enhancing NO
Role of Dysregulated Nutrient Sensing Pathways in bioavailability, and reducing oxidative stress and inflamma-
Vascular Aging: mTOR, Sirtuins, and AMPK tion. Sirtuins are evolutionarily conserved NAD+-dependent
Evolutionarily highly conserved cellular energy-sensing path- protein deacetylases and ADP ribosyltransferases, which
ways were reported to regulate fundamental aging processes regulate multiple pathways involved in the aging process.
858  Circulation Research  September 14, 2018

There is strong evidence that in the vasculature, activation contributes to age-associated endothelial dysfunction. Still,
of members of the sirtuin family, particularly SIRT1, confers little is known regarding the effects of AMPK activators on
multifaceted antiaging effects.42,59,60,66,70,80,143,174–177 SIRT1 spe- other vascular functions, and much more research is needed to
cifically contributes to the vasoprotective effects of calorie better understand the similarities and differences between the
restriction,42,178 augments eNOS activation,143 reduces oxida- effects of various AMPK activators.
tive stress, exerts anti-inflammatory effects,177 is antiapop-
totic, reduces DNA damage, and promotes telomere stability. Role of the RAS in Vascular Aging
SIRT6 also exerts endothelial protective and antiatherogenic In model organisms (Caenorhabditis elegans), reducing the ac-
effects in mice.179,180 In addition to regulating redox homeo- tivity of acn-1 (inactive angiotensin-converting enzyme-related
stasis, mitochondrial function, endothelial vasodilation, protein)—a homologue of the ACE (angiotensin-converting
and protecting against apoptosis and senescence, SIRT1 enzyme)—results in significant extension of life span suggest-
and SIRT6 have been shown to regulate the DNA damage ing that peptide hormones produced by these enzymes regulate
response in VSMCs.181 Studies also suggest that SIRT1 is fundamental aging processes.194 There is initial evidence that
reduced in human atherosclerosis and that SIRT1 exerts an- the antiaging effects of pharmacological ACE inhibitors are
tiatherogenic effects in mouse models by protecting against mediated by pathways that partially overlap with other evo-
DNA damage.181 Importantly, the activity of many sirtuins, lutionarily conserved mechanisms involved in regulation of
including SIRT1 and SIRT6, is dependent on the cellular life span (eg, FOXO [inactive angiotensin-converting enzyme-
NAD+ supply, which is known to decline with age. related protein] signaling194). Studies on laboratory rodents
Because of the multitude of beneficial effects, many extend these findings showing that inhibition of the RAS phar-
sirtuin activators have been utilized to determine whether macologically or by genetic means exerts significant antiaging
sirtuin activation can reverse the vascular aging phenotype. effects, extending life span and reversing age-related phenotyp-
Resveratrol—a naturally occurring polyphenol—can acti- ic and functional changes in the aged vasculature.195–197 There
vate sirtuins and numerous other pathways and has been used is growing evidence demonstrating that upregulation of tissue
extensively to reverse arterial aging.182,183 Additionally, more RAS plays a role in vascular aging promoting intimal thicken-
specific and potent SIRT1 activators, such as SRT1720, have ing and remodeling in large conduit arteries of aged animals
recapitulated the beneficial effects afforded by resveratrol and elderly human subjects.75,198–200 Proof of concept is derived
treatment, demonstrating antioxidant and anti-inflammatory from studies demonstrating that infusion of angiotensin II into
effects. However, although SRT1720 rescued endothelial young rats promotes aging-like changes in the vascular pheno-
function in aged mice, that was the result of enhanced cyclo- type, promoting carotid media thickening and intima infiltra-
Downloaded from http://ahajournals.org by on June 28, 2023

oxegenase vasodilators instead of increased NO bioavailabil- tion by VSMCs.200 Further, pharmacological inhibition of RAS
ity. Supplementation with nicotinamide mononucleotide—a activity can reduce arterial stiffness in aged animals and el-
key NAD+ intermediate—has also been shown increase sir- derly humans independently of changes in blood pressure.201,202
tuin activation,184 resulting in reversal of mitochondrial dys- Upregulation of RAS in the vascular wall likely also promotes
function in advanced age.68 In the aged vasculature, 8 weeks chronic low-grade vascular inflammation and oxidative stress,
of nicotinamide mononucleotide supplementation increases enhancing the vascular response to injury and rendering the
SIRT1 activity and reverses age-related endothelial dysfunc- aged vascular wall susceptible to atherogenesis.75,199 Activation
tion and oxidative stress.70 In summary, SIRT1 and other of RAS/increased angiotensin II levels in aging have also been
sirtuins are considered promising drug targets,177 and future linked to induction of mitochondrial oxidative stress in the vas-
studies should evaluate the efficacy other sirtuin activating culature,203 development of cerebral microhemorrhages,45 and
compounds for cardiovascular protection in older people. disruption of the blood-brain barrier.204
AMPK signaling is an important energy-sensing path- More recently, the view has emerged that an extended
way that is involved in regulation of aging processes, in- renin-angiotensin-aldosterone system, including local ex-
tegrating energy balance, metabolism, and cellular stress pression of mineralocorticoids and their receptors in the
resistance.185,186 In lower organisms, increased AMPK activity vasculature, plays a tissue-specific role in regulation of ag-
was shown to extend life span. Treatment of aged mice with ing processes. It has been well established that circulating
pharmacological activators of AMPK, including metformin or aldosterone regulates water and electrolyte homeostasis and
aminoimidazole carboxamide ribonucleotide, was also shown thereby controls blood pressure. In addition, aldosterone also
to confer significant health benefits. There is currently an ag- promotes structural and functional alterations in the vascula-
ing clinical trial, TAME (Targeting Aging With Metformin), ture, including inflammation and pathological remodeling.205
that aims to determine whether metformin can delay the onset Recent studies demonstrate that age-associated changes of
of age-related diseases in humans.187 In the context of vascu- aldosterone and mineralocorticoid receptor signaling dys-
lar aging, AMPK was shown to confer vasoprotective effects, regulation occur in VSMCs, which may contribute to age-
augmenting eNOS activation.188–191 Furthermore, inhibition of associated arterial remodeling.205 These findings encourage
NF-κB signaling by AMPK suppresses inflammatory process- further experimentation aiming to better characterize the
es. Importantly, AMPK activity is reduced in the aorta192 and pathophysiological relevance of aging-induced alterations
cerebral arteries193 of old rodents. Pharmacological activation in the extended renin-angiotensin-aldosterone system and
of AMPK by aminoimidazole carboxamide ribonucleotide to the efficiency of treatments targeting the extended renin-
was shown to restore endothelium-dependent vasodilation angiotensin-aldosterone system for cardiovascular protection
in old mice,192 suggesting that inactivation of arterial AMPK in older individuals.
Ungvari et al   Mechanisms of Vascular Aging   859

Role of Extracellular Matrix Remodeling in left ventricular remodeling, diastolic dysfunction, and exac-
Vascular Aging erbates atherogenesis. Because of the dilation of conduit ar-
The extracellular matrix (ECM) is an important contribu- teries, wall tension significantly increases, contributing to the
tor to health and longevity. This noncellular compartment, development of aneurysms. In addition to alterations in the
ubiquitous to all tissues and organs does not only provide es- biomechanical properties of large arteries, age-related ECM
sential mechanical scaffolding but mediates highly dynamic remodeling likely also affects microvascular transport and
biomechanical and biochemical signals required for tissue barrier functions.213 Age-related alteration of the ECM struc-
homeostasis, morphogenesis, and cell differentiation. Studies ture and composition are also manifested in the wall of veins,
on model organisms suggest that evolutionarily conserved contributing to the pathogenesis of varicosities.214
pathways regulate ECM remodeling during aging and that
promotion of ECM youthfulness by antiaging interventions Role of Progeronic and Antigeronic Circulating
is an essential signature of longevity assurance.206 Aging in Factors: Lessons Learnt From Heterochronic
mammals also results in significant changes in ECM biosyn- Parabiosis and Caloric Restriction
thesis, postsynthetic modifications of ECM components, and There is growing evidence that noncell-autonomous mecha-
alterations of cell-matrix interactions, which contribute to the nisms play a critical role in orchestrating vascular aging pro-
development of a spectrum of age-related pathologies.207 cesses (Figure 1). Aging-induced alterations in vasoprotective
Age-related alterations of the ECM, including the suben- endocrine factors are of particular importance. Such changes
dothelial basement membrane, intima, media, adventitia, and include an age-related decline in circulating levels of growth
interstitial matrix (which constitute more than half of the mass hormone,215 IGF-1,216 and estrogens, all of which regulate
of the vascular tissue), play a fundamental role in impairment multiple aspects of endothelium-dependent vasodilation,217
of both structural and regulatory homeostasis of the vascula- autoregulation of blood flow,218 vascular structural remodel-
ture.208 With age, the expression of growth factors that regulate ing, atherogenesis,219 and angiogenic processes.220
ECM biosynthesis is altered45 and the synthesis of many ECM The impact of circulating factors on aging phenotypes
components (eg, elastin) declines, which impairs elasticity was also demonstrated by studies using mice with heter-
and resilience of the vascular wall to mechanical damage and ochronic parabiosis, which involves surgically connecting
rupture induced by bursts in wall tension because of pulsatile the circulatory system of a young and an aged mouse.221
pressure waves.208 Age-related ECM changes also likely alter Cerebromicrovascular density typically declines with ad-
vascular mechanotransduction, dysregulating cell responses to vanced age,222 and there is initial evidence that circulating an-
alterations in the hemodynamic environment. Additionally, ag- tigeronic factors (which reverse/prevent development of aging
Downloaded from http://ahajournals.org by on June 28, 2023

ing and cellular senescence alter the secretory phenotype of phenotypes) present in young mice can rejuvenate microvas-
vascular endothelial and smooth muscle cells, increasing MMP cular network architecture in aged heterochronic parabionts.221
secretion.45 This together with increased MMP activation208 in- The antigeronic circulating factors present in young mice
duced by high ROS levels compromises the structural integrity are currently unknown, and the previously proposed role for
of the vasculature and promotes pathological remodeling (eg, GDF11 (growth differentiation factor 11)221 remains contro-
in hypertension), resulting in increased likelihood of aneurysm versial. Future studies should identify additional antigeronic
formation and vessel rupture, including the development of factors that might be targeted by interventions to extend vas-
cerebral microhemorrhages.45 The available evidence suggests cular health span. Progeronic circulating factors increase with
that many of these age-related ECM alterations are governed age and impair tissue homeostasis in young animals. There
by circulating factors and factors produced in the vascular wall, is initial evidence that mediators secreted by senescent cells
including the extended renin-angiotensin-aldosterone system (eg, inflammatory cytokines, such as TNF-α35) may serve as
(see above) and an age-related decline in circulating IGF-1.209 progeronic circulating factors. Further studies are warranted
Collagen synthesis is also dysregulated with age in the to identify additional progeronic proteins and determine their
vascular wall likely because of the effects of increased para- impact on atherogenesis, endothelial function, blood-brain
crine action of TGF-β (transforming growth factor-β),123 barrier integrity, and microvascular function in aging.
which contributes to vascular fibrosis and arterial stiffen- Additional evidence to support a central role of antiger-
ing.208 Additional features that contribute to increased ar- onic circulating factors governing vascular aging processes is
terial stiffness include decreased elastin synthesis, elastin derived from studies on caloric restriction—a dietary regimen,
degradation and fragmentation, elastin calcification, al- which improves health and slow the aging process in evolu-
terations in cross-linking of extracellular matrix compo- tionarily distant organisms.223 Caloric restriction was shown to
nents (eg, by increased presence of advanced glycation end promote a youthful endothelial phenotype by upregulating and
products).208,210,211 activating eNOS in aged animals223–225 and perhaps humans.226
The pathophysiological consequences of age-related A critical role of antigeronic circulating factors in vasculo-
ECM remodeling and arterial stiffening have been the sub- protective phenotypic responses induced by caloric restriction
ject of a recent comprehensive review by AlGhatrif and was first indicated by the observations that in vitro treatment of
Lakatta.6 In brief, as the large conduit arteries stiffen in ag- cultured aged endothelial cells with sera derived from caloric
ing, aortic pulse wave velocity, systolic pressure, and pulse restricted animals mimics phenotypic effects observed in vivo
pressure significantly increase,212 whereas diastolic pressure during caloric restriction, promoting anti-inflammatory and
decreases. Decreased diastolic pressure leads to a decline in proangiogenic effects.42,227 Treatment with sera derived from
coronary blood flow. Increased systolic pressure promotes caloric restricted animals upregulates SIRT1228; however, the
860  Circulation Research  September 14, 2018

exact nature of the circulating factor responsible for this effect Limitations of translating the results of preclinical studies
remains elusive. One potential candidate antigeronic protein should be recognized. An important recent example is caloric
mediating the vasoprotective effects of caloric restriction is restriction.239 Although caloric restriction confers significant
adiponectin, whose serum level is known to be increased by life span extension and cardiovascular protection in labora-
caloric restriction.229–231 tory rodents5,18,42,97,223,240,241 and in certain cohorts of nonhuman
Human studies are needed to identify novel progeronic primates,227,242 its protective effects in nonhuman primates in
and antigeronic circulating factors and their cofactors, acti- other studies243 and in patients with multiple cardiovascular
vators, or inhibitors/antagonists and to seek associations with risk factors are less evident.244 Additionally, in cross-sectional
vascular aging phenotypes. Future studies should also iden- studies, the older groups may represent a selected long-lived
tify cellular origins of circulating progeronic and antigeronic subset of the younger population. There are existing longitudi-
factors that impact vascular aging and characterize pathologi- nal studies in humans (eg, InCHIANTI study) and nonhuman
cal conditions that alter their levels in circulation with aging. primates, and important information related to mechanisms of
Further, mechanistic studies describing the cellular effects of vascular aging could be derived from add-on studies to these
progeronic and antigeronic circulating factors in the vascular existing cohorts.
wall are warranted. Critical areas of vascular aging research include the role
of senescence, epigenetics, stress resilience, inflammation,
Progenitor Cell Exhaustion macromolecular damage, proteostasis, mitochondrial and
On the basis of the stem cell theory of aging, it is postulated metabolic dysfunction, and impaired stem cell biology. The
that the inability of various types of vascular progenitor cells specific roles for cell-autonomous and noncell-autonomous
to continue to replenish the circulatory system with function- mechanisms contributing to vascular aging need to be eluci-
al differentiated endothelial and smooth muscle cells com- dated further. The role of signal transduction pathways linked
promises the biological functions of the aged vasculature. to regulation of cellular energetics in the vascular aging pro-
Importantly, aging impairs neovascularization, which de- cess should be better defined. Future studies should also lead
pends on the function of highly proliferative EPCs. Previous to improved understanding of the role circadian clocks to
studies provide evidence that aging compromises the func- vascular aging. New studies investigating cellular heteroge-
tion of circulating EPCs,232–234 likely by altering the produc- neity in vascular aging are warranted. Stochastic macromo-
tion of factors promoting cell proliferation, migration, and lecular damage leads to regional variability in the presence of
survival (eg, IGF-1), or by enhancing inflammation and oxi- senescent cells, cells with altered metabolism, mitochondrial
dative stress, activating the RAS and promoting senescence. dysfunction, and increased ROS production. Such regional
Downloaded from http://ahajournals.org by on June 28, 2023

There are data demonstrating that sera derived from young variability likely contributes to the focal development of vas-
animals improves the function of cultured EPCs isolated cular pathologies, ranging from atherosclerotic plaques to mi-
from aged rats,235 suggesting a key role for circulating fac- crohemorrhages. Single-cell gene expression analysis should
tors. There are also preclinical data extant suggesting that facilitate better understanding of the pathophysiological role
progressive progenitor cell deficits may contribute to the de- of functional heterogeneity. Finally, how environmental fac-
velopment of atherosclerosis.234 In particular, treatment with tors and lifestyle choice impact the vascular aging processes
bone marrow-derived progenitor cells isolated from young should be better understood.
mice prevents atherosclerosis progression in ApoE−/− mice, The disposable soma theory of aging predicts that age-
whereas treatment with progenitor cells from older mice is related functional decline occurs because of the accumulation
ineffective.234 It should be noted that role of EPCs in the gen- of random macromolecular damage and that the proportion of
esis of vascular aging phenotypes in humans is note well un- energy investment into cellular maintenance and repair pro-
derstood. For example, patients with peripheral artery disease cesses will determine longevity of the organism. Numerous
exhibit low EPC numbers,236 whereas patients with chronic studies investigating cellular and molecular stress responses
myocardial ischemia from coronary microvascular disease237 in the vasculature agree with the predictions of this theory.
or abdominal aorta aneurism have significantly higher levels For example, cells of the long-lived muroid rodent species
of circulating progenitor cells than age‐matched controls.238 white-footed mouse (Peromyscus leucopus; maximum life
Future studies should investigate the synergistic effects of ag- span, >8 years) exhibits decreased production of ROS, im-
ing and associated cardiovascular risk factors in humans and proved antioxidant defense mechanisms, increased resistance
determine how declining EPC function contributes to differ- against oxidative stressors, superior DNA repair mechanisms,
ent vascular aging phenotypes. and more efficient mitochondria than the shorter-lived Mus
musculus.245–247 Similar findings were reported in the vascu-
Future Directions lature of the longest living rodent species—the naked mole
Although significant progress has been achieved in character- rat (Heterocephalus glaber; maximum life span, >30 years).23
izing aging-induced changes in vascular function and pheno- Importantly, virtually all of the mechanistic hypotheses re-
type, research efforts should persist in this direction to develop lated to vascular aging are tested solely in M. musculus (one
innovative strategies based on recent achievements in the biol- of the least successfully aging species), which is potentially a
ogy of aging to improve vascular health span. Understanding source for significant bias and limit the translatability of the
the interactions between aging processes and chronic diseases results to the clinical scenario. Therefore, there is a great need
should be of high priority. Better alignment of preclinical for interspecies comparative studies using animals with dis-
studies on vascular aging and human investigations is needed. parate longevity and human studies to understand generalized
Ungvari et al   Mechanisms of Vascular Aging   861

mechanisms of vascular aging and identify translationally 5. Alfaras I, Di Germanio C, Bernier M, Csiszar A, Ungvari Z, Lakatta EG,
de Cabo R. Pharmacological strategies to retard cardiovascular aging. Circ
relevant treatments for the promotion of vascular health in
Res. 2016;118:1626–1642. doi: 10.1161/CIRCRESAHA.116.307475
older adults. 6. AlGhatrif M, Lakatta EG. The conundrum of arterial stiffness, elevat-
The same cellular and molecular aging processes that af- ed blood pressure, and aging. Curr Hypertens Rep. 2015;17:12. doi:
fect arterial vessels and capillaries also affect veins and the 10.1007/s11906-014-0523-z
7. Wang M, Monticone RE, Lakatta EG. Arterial aging: a journey into sub-
lymphatic/glymphatic system, likely contributing to various clinical arterial disease. Curr Opin Nephrol Hypertens. 2010;19:201–207.
disease pathologies. Examples include the potential role of doi: 10.1097/MNH.0b013e3283361c0b
cerebral venules in neuroinflammation, Alzheimer disease, 8. Seals DR, Desouza CA, Donato AJ, Tanaka H. Habitual exercise
and arterial aging. J Appl Physiol (1985). 2008;105:1323–1332. doi:
and cerebral microhemorrhages46 and the potential link
10.1152/japplphysiol.90553.2008
between age-related glymphatic dysfunction and amyloid 9. Brandes RP, Fleming I, Busse R. Endothelial aging. Cardiovasc Res.
pathologies.248 These areas are attractive targets for future 2005;66:286–294. doi: 10.1016/j.cardiores.2004.12.027
studies. 10. Lacolley P, Regnault V, Avolio AP. Smooth muscle cell and arterial ag-
ing: basic and clinical aspects. Cardiovasc Res. 2018;114:513–528. doi:
Taken together, instead of targeting a single disease, in- 10.1093/cvr/cvy009
terventions that target fundamental aging processes have the 11. Ungvari Z, Kaley G, de Cabo R, Sonntag WE, Csiszar A. Mechanisms
potential to prevent/delay a range of vascular pathologies and of vascular aging: new perspectives. J Gerontol A Biol Sci Med Sci.
2010;65:1028–1041. doi: 10.1093/gerona/glq113
other age-related diseases simultaneously. In recent years, a
12. Harman D. Aging: a theory based on free radical and radiation chemistry.
variety of candidate drugs/interventions have emerged from J Gerontol. 1956;11:298–300.
basic research and translational studies that may target aging 13. Csiszar A, Ungvari Z, Edwards JG, Kaminski P, Wolin MS, Koller A,
processes. These interventions can be adapted for prevention/ Kaley G. Aging-induced phenotypic changes and oxidative stress impair
coronary arteriolar function. Circ Res. 2002;90:1159–1166.
treatment of age-related vascular pathologies. 14. van der Loo B, Labugger R, Skepper JN, Bachschmid M, Kilo J, Powell
JM, Palacios-Callender M, Erusalimsky JD, Quaschning T, Malinski T,
Disclosures Gygi D, Ullrich V, Lüscher TF. Enhanced peroxynitrite formation is as-
sociated with vascular aging. J Exp Med. 2000;192:1731–1744.
None.
15. Adler A, Messina E, Sherman B, Wang Z, Huang H, Linke A, Hintze
TH. NAD(P)H oxidase-generated superoxide anion accounts for re-
Sources of Funding duced control of myocardial O2 consumption by NO in old Fischer 344
This work was supported by grants from the American Heart rats. Am J Physiol Heart Circ Physiol. 2003;285:H1015–H1022. doi:
10.1152/ajpheart.01047.2002
Association (to S. Tarantini), the National Institute on Aging (R01-
16. Donato AJ, Eskurza I, Silver AE, Levy AS, Pierce GL, Gates PE, Seals
AG055395, R01-AG047879, R01-AG038747, 1R01AG057964-01,
DR. Direct evidence of endothelial oxidative stress with aging in hu-
1R21AG055090-01A1, 2AG013319-21, R44-AG053131, K02- mans: relation to impaired endothelium-dependent dilation and upregu-
AG045339, and R01-AG050238), the National Institute of
Downloaded from http://ahajournals.org by on June 28, 2023

lation of nuclear factor-kappaB. Circ Res. 2007;100:1659–1666. doi:


Neurological Disorders and Stroke (R01-NS056218 and R01- 10.1161/01.RES.0000269183.13937.e8
NS100782), Veterans Administration Merit Award 1 I01 BX002211- 17. Jacobson A, Yan C, Gao Q, Rincon-Skinner T, Rivera A, Edwards J, Huang
01A2, the Oklahoma Center for the Advancement of Science and A, Kaley G, Sun D. Aging enhances pressure-induced arterial superoxide
Technology, the Presbyterian Health Foundation, the European formation. Am J Physiol Heart Circ Physiol. 2007;293:H1344–H1350.
Union–funded grant EFOP-3.6.1-16-2016-00008, the Owens doi: 10.1152/ajpheart.00413.2007
Foundation, Kleberg Foundation, San Antonio Medical Foundation, 18. Csiszar A, Gautam T, Sosnowska D, Tarantini S, Banki E, Tucsek Z,
and the Robert L. Bailey and daughter Lisa K. Bailey Alzheimer’s Toth P, Losonczy G, Koller A, Reglodi D, Giles CB, Wren JD, Sonntag
Fund in the memory of Jo Nell Bailey. WE, Ungvari Z. Caloric restriction confers persistent anti-oxidative,
pro-angiogenic, and anti-inflammatory effects and promotes anti-aging
miRNA expression profile in cerebromicrovascular endothelial cells of
Acknowledgments aged rats. Am J Physiol Heart Circ Physiol. 2014;307:H292–H306. doi:
We acknowledge the support from the National Institute on Aging– 10.1152/ajpheart.00307.2014
funded Geroscience Training Program in Oklahoma (T32AG052363). 19. Csiszar A, Sosnowska D, Wang M, Lakatta EG, Sonntag WE, Ungvari Z.
Age-associated proinflammatory secretory phenotype in vascular smooth
muscle cells from the non-human primate Macaca mulatta: reversal by
References resveratrol treatment. J Gerontol A Biol Sci Med Sci. 2012;67:811–820.
1. National Center for Health Statistics. Health, United States, 2016: With doi: 10.1093/gerona/glr228
Chartbook on Long-Term Trends in Health. Hyattsville, MD: National 20. Sun D, Huang A, Yan EH, Wu Z, Yan C, Kaminski PM, Oury TD, Wolin
Center for Health Statistics (US); 2017. MS, Kaley G. Reduced release of nitric oxide to shear stress in mesenteric
2. Heidenreich PA, Trogdon JG, Khavjou OA, et al; American Heart arteries of aged rats. Am J Physiol Heart Circ Physiol. 2004;286:H2249–
Association Advocacy Coordinating Committee; Stroke Council; H2256. doi: 10.1152/ajpheart.00854.2003
Council on Cardiovascular Radiology and Intervention; Council on 21. Hamilton CA, Brosnan MJ, McIntyre M, Graham D, Dominiczak AF.
Clinical Cardiology; Council on Epidemiology and Prevention; Council Superoxide excess in hypertension and aging: a common cause of endo-
on Arteriosclerosis; Thrombosis and Vascular Biology; Council on thelial dysfunction. Hypertension. 2001;37:529–534.
Cardiopulmonary; Critical Care; Perioperative and Resuscitation; 22. Francia P, delli Gatti C, Bachschmid M, Martin-Padura I, Savoia C,
Council on Cardiovascular Nursing; Council on the Kidney in Migliaccio E, Pelicci PG, Schiavoni M, Lüscher TF, Volpe M, Cosentino
Cardiovascular Disease; Council on Cardiovascular Surgery and F. Deletion of p66shc gene protects against age-related endothelial dys-
Anesthesia, and Interdisciplinary Council on Quality of Care and function. Circulation. 2004;110:2889–2895. doi: 10.1161/01.CIR.
Outcomes Research. Forecasting the future of cardiovascular dis- 0000147731.24444.4D
ease in the United States: a policy statement from the American Heart 23. Csiszar A, Labinskyy N, Orosz Z, Xiangmin Z, Buffenstein R,

Association. Circulation. 2011;123:933–944. doi: 10.1161/CIR. Ungvari Z. Vascular aging in the longest-living rodent, the naked mole
0b013e31820a55f5 rat. Am J Physiol Heart Circ Physiol. 2007;293:H919–H927. doi:
3. Lakatta EG, Levy D. Arterial and cardiac aging: major shareholders in 10.1152/ajpheart.01287.2006
cardiovascular disease enterprises: part I: aging arteries: a “set up” for 24. Ungvari Z, Orosz Z, Labinskyy N, Rivera A, Xiangmin Z, Smith K,
vascular disease. Circulation. 2003;107:139–146. Csiszar A. Increased mitochondrial H2O2 production promotes endothe-
4. López-Otín C, Blasco MA, Partridge L, Serrano M, Kroemer G. The hall- lial NF-kappaB activation in aged rat arteries. Am J Physiol Heart Circ
marks of aging. Cell. 2013;153:1194–1217. doi: 10.1016/j.cell.2013.05.039 Physiol. 2007;293:H37–H47. doi: 10.1152/ajpheart.01346.2006
862  Circulation Research  September 14, 2018

25. Jablonski KL, Seals DR, Eskurza I, Monahan KD, Donato AJ. High- 45. Toth P, Tarantini S, Springo Z, Tucsek Z, Gautam T, Giles CB, Wren
dose ascorbic acid infusion abolishes chronic vasoconstriction and re- JD, Koller A, Sonntag WE, Csiszar A, Ungvari Z. Aging exacerbates
stores resting leg blood flow in healthy older men. J Appl Physiol (1985). hypertension-induced cerebral microhemorrhages in mice: role of res-
2007;103:1715–1721. doi: 10.1152/japplphysiol.00533.2007 veratrol treatment in vasoprotection. Aging Cell. 2015;14:400–408. doi:
26. Donato AJ, Gano LB, Eskurza I, Silver AE, Gates PE, Jablonski K, 10.1111/acel.12315
Seals DR. Vascular endothelial dysfunction with aging: endothelin-1 46. Ungvari Z, Tarantini S, Kirkpatrick AC, Csiszar A, Prodan CI.

and endothelial nitric oxide synthase. Am J Physiol Heart Circ Physiol. Cerebral microhemorrhages: mechanisms, consequences, and preven-
2009;297:H425–H432. doi: 10.1152/ajpheart.00689.2008 tion. Am J Physiol Heart Circ Physiol. 2017;312:H1128–H1143. doi:
27. Spier SA, Delp MD, Meininger CJ, Donato AJ, Ramsey MW, Muller-Delp 10.1152/ajpheart.00780.2016
JM. Effects of ageing and exercise training on endothelium-dependent 47. Fleenor BS, Eng JS, Sindler AL, Pham BT, Kloor JD, Seals DR.

vasodilatation and structure of rat skeletal muscle arterioles. J Physiol. Superoxide signaling in perivascular adipose tissue promotes age-related
2004;556:947–958. doi: 10.1113/jphysiol.2003.060301 artery stiffness. Aging Cell. 2014;13:576–578. doi: 10.1111/acel.12196
28. Durrant JR, Seals DR, Connell ML, Russell MJ, Lawson BR, Folian BJ, 48. Wallace DC. A mitochondrial paradigm of metabolic and degenerative
Donato AJ, Lesniewski LA. Voluntary wheel running restores endothe- diseases, aging, and cancer: a dawn for evolutionary medicine. Annu Rev
lial function in conduit arteries of old mice: direct evidence for reduced Genet. 2005;39:359–407. doi: 10.1146/annurev.genet.39.110304.095751
oxidative stress, increased superoxide dismutase activity and down- 49. Schriner SE, Linford NJ, Martin GM, Treuting P, Ogburn CE, Emond
regulation of NADPH oxidase. J Physiol. 2009;587:3271–3285. doi: M, Coskun PE, Ladiges W, Wolf N, Van Remmen H, Wallace DC,
10.1113/jphysiol.2009.169771 Rabinovitch PS. Extension of murine life span by overexpression of
29. Novella S, Dantas AP, Segarra G, Novensa L, Heras M, Hermenegildo C, catalase targeted to mitochondria. Science. 2005;308:1909–1911. doi:
Medina P. Aging enhances contraction to thromboxane A2 in aorta from 10.1126/science.1106653
female senescence-accelerated mice. Age (Dordr). 2013;35:117–128. doi: 50. Springo Z, Tarantini S, Toth P, Tucsek Z, Koller A, Sonntag WE, Csiszar
10.1007/s11357-011-9337-y A, Ungvari Z. Aging exacerbates pressure-induced mitochondrial oxi-
30. Cardillo C, Kilcoyne CM, Cannon RO III, Panza JA. Interactions between dative stress in mouse cerebral arteries. J Gerontol A Biol Sci Med Sci.
nitric oxide and endothelin in the regulation of vascular tone of human 2015;70:1355–1359. doi: 10.1093/gerona/glu244
resistance vessels in vivo. Hypertension. 2000;35:1237–1241. 51. Tarantini S, Valcarcel-Ares NM, Yabluchanskiy A, Fulop GA, Hertelendy
31. Park L, Anrather J, Girouard H, Zhou P, Iadecola C. Nox2-derived re- P, Gautam T, Farkas E, Perz A, Rabinovitch PS, Sonntag WE, Csiszar A,
active oxygen species mediate neurovascular dysregulation in the ag- Ungvari Z. Treatment with the mitochondrial-targeted antioxidant peptide
ing mouse brain. J Cereb Blood Flow Metab. 2007;27:1908–1918. doi: SS-31 rescues neurovascular coupling responses and cerebrovascular en-
10.1038/sj.jcbfm.9600491 dothelial function and improves cognition in aged mice [published online
32. Toth P, Tarantini S, Tucsek Z, Ashpole NM, Sosnowska D, Gautam T, February 6, 2018]. Aging Cell. doi: 10.1111/acel.12731
Ballabh P, Koller A, Sonntag WE, Csiszar A, Ungvari Z. Resveratrol treat- 52. Ungvari Z, Labinskyy N, Gupte S, Chander PN, Edwards JG, Csiszar
ment rescues neurovascular coupling in aged mice: role of improved ce- A. Dysregulation of mitochondrial biogenesis in vascular endothelial
rebromicrovascular endothelial function and downregulation of NADPH and smooth muscle cells of aged rats. Am J Physiol Heart Circ Physiol.
oxidase. Am J Physiol Heart Circ Physiol. 2014;306:H299–H308. doi: 2008;294:H2121–H2128. doi: 10.1152/ajpheart.00012.2008
10.1152/ajpheart.00744.2013 53. Napoli C, Martin-Padura I, de Nigris F, Giorgio M, Mansueto G, Somma
33. Cernadas MR, Sánchez de Miguel L, García-Durán M, González-
P, Condorelli M, Sica G, De Rosa G, Pelicci P. Deletion of the p66Shc
Fernández F, Millás I, Montón M, Rodrigo J, Rico L, Fernández P, de longevity gene reduces systemic and tissue oxidative stress, vascular cell
Downloaded from http://ahajournals.org by on June 28, 2023

Frutos T, Rodríguez-Feo JA, Guerra J, Caramelo C, Casado S, López- apoptosis, and early atherogenesis in mice fed a high-fat diet. Proc Natl
Farré. Expression of constitutive and inducible nitric oxide synthases in Acad Sci USA. 2003;100:2112–2116. doi: 10.1073/pnas.0336359100
the vascular wall of young and aging rats. Circ Res. 1998;83:279–286. 54. Ungvari Z, Bailey-Downs L, Gautam T, Sosnowska D, Wang M, Monticone
34. Chou TC, Yen MH, Li CY, Ding YA. Alterations of nitric oxide syn- RE, Telljohann R, Pinto JT, de Cabo R, Sonntag WE, Lakatta EG, Csiszar
thase expression with aging and hypertension in rats. Hypertension. A. Age-associated vascular oxidative stress, Nrf2 dysfunction, and NF-
1998;31:643–648. {kappa}B activation in the nonhuman primate Macaca mulatta. J Gerontol
35. Csiszar A, Labinskyy N, Smith K, Rivera A, Orosz Z, Ungvari Z.
A Biol Sci Med Sci. 2011;66:866–875. doi: 10.1093/gerona/glr092
Vasculoprotective effects of anti-tumor necrosis factor-alpha treatment in 55. Ungvari Z, Bailey-Downs L, Sosnowska D, Gautam T, Koncz P, Losonczy
aging. Am J Pathol. 2007;170:388–398. doi: 10.2353/ajpath.2007.060708 G, Ballabh P, de Cabo R, Sonntag WE, Csiszar A. Vascular oxidative stress
36. Ganz P, Vita JA. Testing endothelial vasomotor function: nitric ox-
in aging: a homeostatic failure due to dysregulation of NRF2-mediated
ide, a multipotent molecule. Circulation. 2003;108:2049–2053. doi: antioxidant response. Am J Physiol Heart Circ Physiol. 2011;301:H363–
10.1161/01.CIR.0000089507.19675.F9 H372. doi: 10.1152/ajpheart.01134.2010
37. Moncada S, Higgs A. The L-arginine-nitric oxide pathway. N Engl J Med. 56. Burns EM, Kruckeberg TW, Comerford LE, Buschmann MT. Thinning
1993;329:2002–2012. doi: 10.1056/NEJM199312303292706 of capillary walls and declining numbers of endothelial mitochondria in
38. Moncada S, Palmer RM, Higgs EA. Nitric oxide: physiology, pathophysi- the cerebral cortex of the aging primate, Macaca nemestrina. J Gerontol.
ology, and pharmacology. Pharmacol Rev. 1991;43:109–142. 1979;34:642–650.
39. Pacher P, Beckman JS, Liaudet L. Nitric oxide and peroxynitrite in
57. Burns EM, Kruckeberg TW, Gaetano PK. Changes with age in cerebral
health and disease. Physiol Rev. 2007;87:315–424. doi: 10.1152/ capillary morphology. Neurobiol Aging. 1981;2:283–291.
physrev.00029.2006 58. Gioscia-Ryan RA, LaRocca TJ, Sindler AL, Zigler MC, Murphy MP, Seals
40. Csiszar A, Podlutsky A, Wolin MS, Losonczy G, Pacher P, Ungvari Z. DR. Mitochondria-targeted antioxidant (MitoQ) ameliorates age-related
Oxidative stress and accelerated vascular aging: implications for cigarette arterial endothelial dysfunction in mice. J Physiol. 2014;592:2549–2561.
smoking. Front Biosci (Landmark Ed). 2009;14:3128–3144. doi: 10.1113/jphysiol.2013.268680
41. Csiszar A, Wang M, Lakatta EG, Ungvari Z. Inflammation and endothe- 59. Pearson KJ, Baur JA, Lewis KN, et al. Resveratrol delays age-related
lial dysfunction during aging: role of NF-kappaB. J Appl Physiol (1985). deterioration and mimics transcriptional aspects of dietary restric-
2008;105:1333–1341. doi: 10.1152/japplphysiol.90470.2008 tion without extending life span. Cell Metab. 2008;8:157–168. doi:
42. Csiszar A, Labinskyy N, Jimenez R, Pinto JT, Ballabh P, Losonczy G, 10.1016/j.cmet.2008.06.011
Pearson KJ, de Cabo R, Ungvari Z. Anti-oxidative and anti-inflamma- 60. Ungvari Z, Labinskyy N, Mukhopadhyay P, Pinto JT, Bagi Z, Ballabh P,
tory vasoprotective effects of caloric restriction in aging: role of circu- Zhang C, Pacher P, Csiszar A. Resveratrol attenuates mitochondrial oxida-
lating factors and SIRT1. Mech Ageing Dev. 2009;130:518–527. doi: tive stress in coronary arterial endothelial cells. Am J Physiol Heart Circ
10.1016/j.mad.2009.06.004 Physiol. 2009;297:H1876–H1881. doi: 10.1152/ajpheart.00375.2009
43. Moreau KL, Gavin KM, Plum AE, Seals DR. Ascorbic acid selectively 61. Rice KM, Preston DL, Walker EM, Blough ER. Aging influences mul-
improves large elastic artery compliance in postmenopausal women. tiple incidices of oxidative stress in the aortic media of the Fischer 344/
Hypertension. 2005;45:1107–1112. doi: 10.1161/01.HYP.0000165678. NNiaxBrown Norway/BiNia rat. Free Radic Res. 2006;40:185–197. doi:
63373.8c 10.1080/10715760500464957
44. Kaneko H, Anzai T, Morisawa M, et al. Resveratrol prevents the develop- 62. Wenzel P, Schuhmacher S, Kienhöfer J, Müller J, Hortmann M, Oelze M,
ment of abdominal aortic aneurysm through attenuation of inflammation, Schulz E, Treiber N, Kawamoto T, Scharffetter-Kochanek K, Münzel T,
oxidative stress, and neovascularization. Atherosclerosis. 2011;217:350– Bürkle A, Bachschmid MM, Daiber A. Manganese superoxide dismutase
357. doi: 10.1016/j.atherosclerosis.2011.03.042 and aldehyde dehydrogenase deficiency increase mitochondrial oxidative
Ungvari et al   Mechanisms of Vascular Aging   863

stress and aggravate age-dependent vascular dysfunction. Cardiovasc Res. of a senescence-associated secretory phenotype and impairs angiogenic
2008;80:280–289. doi: 10.1093/cvr/cvn182 capacity in cerebromicrovascular endothelial cells: role of increased
63. Botto N, Berti S, Manfredi S, Al-Jabri A, Federici C, Clerico A, Ciofini E, DNA damage and decreased DNA repair capacity in microvascular ra-
Biagini A, Andreassi MG. Detection of mtDNA with 4977 bp deletion in diosensitivity. J Gerontol A Biol Sci Med Sci. 2013;68:1443–1457. doi:
blood cells and atherosclerotic lesions of patients with coronary artery dis- 10.1093/gerona/glt057
ease. Mutat Res. 2005;570:81–88. doi: 10.1016/j.mrfmmm.2004.10.003 82. Freund A, Patil CK, Campisi J. p38MAPK is a novel DNA damage re-
64. Bogliolo M, Izzotti A, De Flora S, Carli C, Abbondandolo A, Degan P. sponse-independent regulator of the senescence-associated secretory phe-
Detection of the ‘4977 bp’ mitochondrial DNA deletion in human athero- notype. EMBO J. 2011;30:1536–1548. doi: 10.1038/emboj.2011.69
sclerotic lesions. Mutagenesis. 1999;14:77–82. 83. Kang C, Xu Q, Martin TD, Li MZ, Demaria M, Aron L, Lu T, Yankner
65. Yu E, Calvert PA, Mercer JR, et al. Mitochondrial DNA damage can pro- BA, Campisi J, Elledge SJ. The DNA damage response induces inflam-
mote atherosclerosis independently of reactive oxygen species through mation and senescence by inhibiting autophagy of GATA4. Science.
effects on smooth muscle cells and monocytes and correlates with 2015;349:aaa5612. doi: 10.1126/science.aaa5612
higher-risk plaques in humans. Circulation. 2013;128:702–712. doi: 84. Morgan RG, Ives SJ, Lesniewski LA, Cawthon RM, Andtbacka RH,
10.1161/CIRCULATIONAHA.113.002271 Noyes RD, Richardson RS, Donato AJ. Age-related telomere uncapping
66. Csiszar A, Labinskyy N, Pinto JT, Ballabh P, Zhang H, Losonczy G, is associated with cellular senescence and inflammation independent of
Pearson K, de Cabo R, Pacher P, Zhang C, Ungvari Z. Resveratrol induces telomere shortening in human arteries. Am J Physiol Heart Circ Physiol.
mitochondrial biogenesis in endothelial cells. Am J Physiol Heart Circ 2013;305:H251–H258. doi: 10.1152/ajpheart.00197.2013
Physiol. 2009;297:H13–H20. doi: 10.1152/ajpheart.00368.2009 85. Youm YH, Grant RW, McCabe LR, Albarado DC, Nguyen KY, Ravussin
67. Lee IH, Cao L, Mostoslavsky R, Lombard DB, Liu J, Bruns NE, Tsokos A, Pistell P, Newman S, Carter R, Laque A, Münzberg H, Rosen CJ,
M, Alt FW, Finkel T. A role for the NAD-dependent deacetylase Sirt1 in Ingram DK, Salbaum JM, Dixit VD. Canonical Nlrp3 inflammasome
the regulation of autophagy. Proc Natl Acad Sci USA. 2008;105:3374– links systemic low-grade inflammation to functional decline in aging. Cell
3379. doi: 10.1073/pnas.0712145105 Metab. 2013;18:519–532. doi: 10.1016/j.cmet.2013.09.010
68. Gomes AP, Price NL, Ling AJ, et al. Declining NAD(+) induces a
86. Bailey-Downs LC, Tucsek Z, Toth P, Sosnowska D, Gautam T, Sonntag
pseudohypoxic state disrupting nuclear-mitochondrial communica- WE, Csiszar A, Ungvari Z. Aging exacerbates obesity-induced oxida-
tion during aging. Cell. 2013;155:1624–1638. doi: 10.1016/j.cell. tive stress and inflammation in perivascular adipose tissue in mice: a
2013.11.037 paracrine mechanism contributing to vascular redox dysregulation and
69. Massudi H, Grant R, Braidy N, Guest J, Farnsworth B, Guillemin GJ. Age- inflammation. J Gerontol A Biol Sci Med Sci. 2013;68:780–792. doi:
associated changes in oxidative stress and NAD+ metabolism in human 10.1093/gerona/gls238
tissue. PLoS One. 2012;7:e42357. doi: 10.1371/journal.pone.0042357 87. Tucsek Z, Toth P, Sosnowska D, Gautam T, Mitschelen M, Koller A,
70. de Picciotto NE, Gano LB, Johnson LC, Martens CR, Sindler AL, Mills Szalai G, Sonntag WE, Ungvari Z, Csiszar A. Obesity in aging exacerbates
KF, Imai S, Seals DR. Nicotinamide mononucleotide supplementation re- blood-brain barrier disruption, neuroinflammation, and oxidative stress in
verses vascular dysfunction and oxidative stress with aging in mice. Aging the mouse hippocampus: effects on expression of genes involved in beta-
Cell. 2016;15:522–530. doi: 10.1111/acel.12461 amyloid generation and Alzheimer’s disease. J Gerontol A Biol Sci Med
71. Mills KF, Yoshida S, Stein LR, Grozio A, Kubota S, Sasaki Y, Redpath Sci. 2014;69:1212–1226. doi: 10.1093/gerona/glt177
P, Migaud ME, Apte RS, Uchida K, Yoshino J, Imai SI. Long-term ad- 88. Tucsek Z, Toth P, Tarantini S, Sosnowska D, Gautam T, Warrington JP,
ministration of nicotinamide mononucleotide mitigates age-associated Giles CB, Wren JD, Koller A, Ballabh P, Sonntag WE, Ungvari Z, Csiszar
physiological decline in mice. Cell Metab. 2016;24:795–806. doi: A. Aging exacerbates obesity-induced cerebromicrovascular rarefaction,
Downloaded from http://ahajournals.org by on June 28, 2023

10.1016/j.cmet.2016.09.013 neurovascular uncoupling, and cognitive decline in mice. J Gerontol A


72. Franceschi C, Bonafè M, Valensin S, Olivieri F, De Luca M, Ottaviani E, Biol Sci Med Sci. 2014;69:1339–1352. doi: 10.1093/gerona/glu080
De Benedictis G. Inflamm-aging. An evolutionary perspective on immu- 89. Orosz Z, Csiszar A, Labinskyy N, Smith K, Kaminski PM, Ferdinandy P,
nosenescence. Ann NY Acad Sci. 2000;908:244–254. Wolin MS, Rivera A, Ungvari Z. Cigarette smoke-induced proinflamma-
73. Csiszar A, Ungvari Z, Koller A, Edwards JG, Kaley G. Aging-induced tory alterations in the endothelial phenotype: role of NAD(P)H oxidase
proinflammatory shift in cytokine expression profile in coronary arteries. activation. Am J Physiol Heart Circ Physiol. 2007;292:H130–H139. doi:
FASEB J. 2003;17:1183–1185. doi: 10.1096/fj.02-1049fje 10.1152/ajpheart.00599.2006
74. Csiszar A, Ungvari Z, Koller A, Edwards JG, Kaley G. Proinflammatory phe- 90. Staras SA, Dollard SC, Radford KW, Flanders WD, Pass RF, Cannon MJ.
notype of coronary arteries promotes endothelial apoptosis in aging. Physiol Seroprevalence of cytomegalovirus infection in the United States, 1988-
Genomics. 2004;17:21–30. doi: 10.1152/physiolgenomics.00136.2003 1994. Clin Infect Dis. 2006;43:1143–1151. doi: 10.1086/508173
75. Wang M, Zhang J, Jiang LQ, Spinetti G, Pintus G, Monticone R, Kolodgie 91. Wang GC, Kao WH, Murakami P, Xue QL, Chiou RB, Detrick B, McDyer
FD, Virmani R, Lakatta EG. Proinflammatory profile within the grossly JF, Semba RD, Casolaro V, Walston JD, Fried LP. Cytomegalovirus infec-
normal aged human aortic wall. Hypertension. 2007;50:219–227. doi: tion and the risk of mortality and frailty in older women: a prospective
10.1161/HYPERTENSIONAHA.107.089409 observational cohort study. Am J Epidemiol. 2010;171:1144–1152. doi:
76. Song Y, Shen H, Schenten D, Shan P, Lee PJ, Goldstein DR. Aging 10.1093/aje/kwq062
enhances the basal production of IL-6 and CCL2 in vascular smooth 92. Ungvari Z, Bailey-Downs L, Gautam T, Jimenez R, Losonczy G, Zhang
muscle cells. Arterioscler Thromb Vasc Biol. 2012;32:103–109. doi: C, Ballabh P, Recchia FA, Wilkerson DC, Sonntag WE, Pearson K, de
10.1161/ATVBAHA.111.236349 Cabo R, Csiszar A. Adaptive induction of NF-E2-related factor-2-driv-
77. Arenas IA, Xu Y, Davidge ST. Age-associated impairment in vasorelaxation en antioxidant genes in endothelial cells in response to hyperglyce-
to fluid shear stress in the female vasculature is improved by TNF-alpha mia. Am J Physiol Heart Circ Physiol. 2011;300:H1133–H1140. doi:
antagonism. Am J Physiol Heart Circ Physiol. 2006;290:H1259–H1263. 10.1152/ajpheart.00402.2010
doi: 10.1152/ajpheart.00990.2005 93. Suh JH, Shenvi SV, Dixon BM, Liu H, Jaiswal AK, Liu RM, Hagen TM.
78. Hasegawa Y, Saito T, Ogihara T, Ishigaki Y, Yamada T, Imai J, Uno K, Decline in transcriptional activity of Nrf2 causes age-related loss of gluta-
Gao J, Kaneko K, Shimosawa T, Asano T, Fujita T, Oka Y, Katagiri H. thione synthesis, which is reversible with lipoic acid. Proc Natl Acad Sci
Blockade of the nuclear factor-κB pathway in the endothelium prevents USA. 2004;101:3381–3386. doi: 10.1073/pnas.0400282101
insulin resistance and prolongs life spans. Circulation. 2012;125:1122– 94. Mostoslavsky R, Chua KF, Lombard DB, et al. Genomic instabil-

1133. doi: 10.1161/CIRCULATIONAHA.111.054346 ity and aging-like phenotype in the absence of mammalian SIRT6. Cell.
79. Chen HZ, Wang F, Gao P, et al. Age-associated sirtuin 1 reduction in 2006;124:315–329. doi: 10.1016/j.cell.2005.11.044
vascular smooth muscle links vascular senescence and inflammation 95. Valcarcel-Ares MN, Gautam T, Warrington JP, Bailey-Downs L,

to abdominal aortic aneurysm. Circ Res. 2016;119:1076–1088. doi: Sosnowska D, de Cabo R, Losonczy G, Sonntag WE, Ungvari Z, Csiszar
10.1161/CIRCRESAHA.116.308895 A. Disruption of Nrf2 signaling impairs angiogenic capacity of endothelial
80. Gano LB, Donato AJ, Pasha HM, Hearon CM Jr, Sindler AL, Seals DR. cells: implications for microvascular aging. J Gerontol A Biol Sci Med Sci.
The SIRT1 activator SRT1720 reverses vascular endothelial dysfunc- 2012;67:821–829. doi: 10.1093/gerona/glr229
tion, excessive superoxide production, and inflammation with aging in 96. Tarantini S, Valcarcel-Ares MN, Yabluchanskiy A, et al. Nrf2 deficiency
mice. Am J Physiol Heart Circ Physiol. 2014;307:H1754–H1763. doi: exacerbates obesity-induced oxidative stress, neurovascular dysfunction,
10.1152/ajpheart.00377.2014 blood brain barrier disruption, neuroinflammation, amyloidogenic gene
81. Ungvari Z, Podlutsky A, Sosnowska D, Tucsek Z, Toth P, Deak F, Gautam expression and cognitive decline in mice, mimicking the aging phenotype.
T, Csiszar A, Sonntag WE. Ionizing radiation promotes the acquisition J Gerontol A Biol Sci Med Sci. 2018;73:853–863.
864  Circulation Research  September 14, 2018

97. Pearson KJ, Lewis KN, Price NL, et al. Nrf2 mediates cancer protection mice with low levels of BubR1. Stroke. 2007;38:1050–1056. doi:
but not prolongevity induced by caloric restriction. Proc Natl Acad Sci 10.1161/01.STR.0000257967.86132.01
USA. 2008;105:2325–2330. doi: 10.1073/pnas.0712162105 118. Martinet W, Knaapen MW, De Meyer GR, Herman AG, Kockx MM.
98. Kaushik S, Cuervo AM. Proteostasis and aging. Nat Med. 2015;21:1406– Elevated levels of oxidative DNA damage and DNA repair enzymes in
1415. doi: 10.1038/nm.4001 human atherosclerotic plaques. Circulation. 2002;106:927–932.
99. Blice-Baum AC, Zambon AC, Kaushik G, Viswanathan MC, Engler AJ, 119. Gray K, Kumar S, Figg N, Harrison J, Baker L, Mercer J,

Bodmer R, Cammarato A. Modest overexpression of FOXO maintains Littlewood T, Bennett M. Effects of DNA damage in smooth mus-
cardiac proteostasis and ameliorates age-associated functional decline. cle cells in atherosclerosis. Circ Res. 2015;116:816–826. doi:
Aging Cell. 2017;16:93–103. doi: 10.1111/acel.12543 10.1161/CIRCRESAHA.116.304921
100. Dai DF, Karunadharma PP, Chiao YA, Basisty N, Crispin D, Hsieh 120. Olive M, Harten I, Mitchell R, et al. Cardiovascular pathology in

EJ, Chen T, Gu H, Djukovic D, Raftery D, Beyer RP, MacCoss MJ, Hutchinson-Gilford progeria: correlation with the vascular pathology
Rabinovitch PS. Altered proteome turnover and remodeling by short- of aging. Arterioscler Thromb Vasc Biol. 2010;30:2301–2309. doi:
term caloric restriction or rapamycin rejuvenate the aging heart. Aging 10.1161/ATVBAHA.110.209460
Cell. 2014;13:529–539. doi: 10.1111/acel.12203 121. Varga R, Eriksson M, Erdos MR, et al. Progressive vascular smooth
101. Gouveia M, Xia K, Colón W, Vieira SI, Ribeiro F. Protein aggrega- muscle cell defects in a mouse model of Hutchinson-Gilford proge-
tion, cardiovascular diseases, and exercise training: where do we stand? ria syndrome. Proc Natl Acad Sci USA. 2006;103:3250–3255. doi:
Ageing Res Rev. 2017;40:1–10. doi: 10.1016/j.arr.2017.07.005 10.1073/pnas.0600012103
102. Chin JH, Okazaki M, Hu ZW, Miller JW, Hoffman BB. Activation of heat 122. Ragnauth CD, Warren DT, Liu Y, McNair R, Tajsic T, Figg N, Shroff R,
shock protein (hsp)70 and proto-oncogene expression by alpha1 adrener- Skepper J, Shanahan CM. Prelamin A acts to accelerate smooth muscle cell
gic agonist in rat aorta with age. J Clin Invest. 1996;97:2316–2323. doi: senescence and is a novel biomarker of human vascular aging. Circulation.
10.1172/JCI118674 2010;121:2200–2210. doi: 10.1161/CIRCULATIONAHA.109.902056
103. Nussenzweig SC, Verma S, Finkel T. The role of autophagy in vascu- 123. Morgan RG, Ives SJ, Walker AE, Cawthon RM, Andtbacka RH, Noyes
lar biology. Circ Res. 2015;116:480–488. doi: 10.1161/CIRCRESAHA. D, Lesniewski LA, Richardson RS, Donato AJ. Role of arterial telomere
116.303805 dysfunction in hypertension: relative contributions of telomere short-
104. Bharath LP, Mueller R, Li Y, Ruan T, Kunz D, Goodrich R, Mills T, Deeter ening and telomere uncapping. J Hypertens. 2014;32:1293–1299. doi:
L, Sargsyan A, Anandh Babu PV, Graham TE, Symons JD. Impairment 10.1097/HJH.0000000000000157
of autophagy in endothelial cells prevents shear-stress-induced increases 124. Erusalimsky JD. Vascular endothelial senescence: from mechanisms

in nitric oxide bioavailability. Can J Physiol Pharmacol. 2014;92:605– to pathophysiology. J Appl Physiol (1985). 2009;106:326–332. doi:
612. doi: 10.1139/cjpp-2014-0017 10.1152/japplphysiol.91353.2008
105. LaRocca TJ, Henson GD, Thorburn A, Sindler AL, Pierce GL, Seals DR. 125. Baker DJ, Childs BG, Durik M, Wijers ME, Sieben CJ, Zhong J, Saltness
Translational evidence that impaired autophagy contributes to arterial age- RA, Jeganathan KB, Verzosa GC, Pezeshki A, Khazaie K, Miller JD,
ing. J Physiol. 2012;590:3305–3316. doi: 10.1113/jphysiol.2012.229690 van Deursen JM. Naturally occurring p16(Ink4a)-positive cells shorten
106. LaRocca TJ, Gioscia-Ryan RA, Hearon CM Jr, Seals DR. The autoph- healthy lifespan. Nature. 2016;530:184–189. doi: 10.1038/nature16932
agy enhancer spermidine reverses arterial aging. Mech Ageing Dev. 126. Jeon OH, Kim C, Laberge RM, Demaria M, Rathod S, Vasserot AP,
2013;134:314–320. doi: 10.1016/j.mad.2013.04.004 Chung JW, Kim DH, Poon Y, David N, Baker DJ, van Deursen JM,
107. Marfella R, Di Filippo C, Laieta MT, Vestini R, Barbieri M, Sangiulo Campisi J, Elisseeff JH. Local clearance of senescent cells attenuates the
P, Crescenzi B, Ferraraccio F, Rossi F, D’Amico M, Paolisso G. Effects development of post-traumatic osteoarthritis and creates a pro-regenera-
Downloaded from http://ahajournals.org by on June 28, 2023

of ubiquitin-proteasome system deregulation on the vascular senescence tive environment. Nat Med. 2017;23:775–781. doi: 10.1038/nm.4324
and atherosclerosis process in elderly patients. J Gerontol A Biol Sci Med 127. Roos CM, Zhang B, Palmer AK, Ogrodnik MB, Pirtskhalava T, Thalji
Sci. 2008;63:200–203. NM, Hagler M, Jurk D, Smith LA, Casaclang-Verzosa G, Zhu Y, Schafer
108. Bulteau AL, Szweda LI, Friguet B. Age-dependent declines in protea- MJ, Tchkonia T, Kirkland JL, Miller JD. Chronic senolytic treatment
some activity in the heart. Arch Biochem Biophys. 2002;397:298–304. alleviates established vasomotor dysfunction in aged or atherosclerotic
doi: 10.1006/abbi.2001.2663 mice. Aging Cell. 2016;15:973–977. doi: 10.1111/acel.12458
109. Wilck N, Ludwig A. Targeting the ubiquitin-proteasome system in ath- 128. Bhayadia R, Schmidt BM, Melk A, Hömme M. Senescence-induced oxi-
erosclerosis: status quo, challenges, and perspectives. Antioxid Redox dative stress causes endothelial dysfunction. J Gerontol A Biol Sci Med
Signal. 2014;21:2344–2363. doi: 10.1089/ars.2013.5805 Sci. 2016;71:161–169. doi: 10.1093/gerona/glv008
110. Szilard L. On the nature of the aging process. Proc Natl Acad Sci USA. 129. Rossman MJ, Kaplon RE, Hill SD, McNamara MN, Santos-Parker

1959;45:30–45. JR, Pierce GL, Seals DR, Donato AJ. Endothelial cell senescence with
111. Moskalev AA, Shaposhnikov MV, Plyusnina EN, Zhavoronkov A,
aging in healthy humans: prevention by habitual exercise and relation
Budovsky A, Yanai H, Fraifeld VE. The role of DNA damage and to vascular endothelial function. Am J Physiol Heart Circ Physiol.
repair in aging through the prism of Koch-like criteria. Ageing Res Rev. 2017;313:H890–H895. doi: 10.1152/ajpheart.00416.2017
2013;12:661–684. doi: 10.1016/j.arr.2012.02.001 130. Gevaert AB, Lemmens K, Vrints CJ, Van Craenenbroeck EM. Targeting
112. Shah AV, Bennett MR. DNA damage-dependent mechanisms of age- endothelial function to treat heart failure with preserved ejection
ing and disease in the macro- and microvasculature. Eur J Pharmacol. fraction: the promise of exercise training. Oxid Med Cell Longev.
2017;816:116–128. doi: 10.1016/j.ejphar.2017.03.050 2017;2017:4865756. doi: 10.1155/2017/4865756
113. Ashpole NM, Warrington JP, Mitschelen MC, Yan H, Sosnowska D, 131. Childs BG, Baker DJ, Wijshake T, Conover CA, Campisi J, van Deursen
Gautam T, Farley JA, Csiszar A, Ungvari Z, Sonntag WE. Systemic JM. Senescent intimal foam cells are deleterious at all stages of athero-
influences contribute to prolonged microvascular rarefaction after sclerosis. Science. 2016;354:472–477. doi: 10.1126/science.aaf6659
brain irradiation: a role for endothelial progenitor cells. Am J Physiol 132. Gardner SE, Humphry M, Bennett MR, Clarke MC. Senescent vascu-
Heart Circ Physiol. 2014;307:H858–H868. doi: 10.1152/ajpheart. lar smooth muscle cells drive inflammation through an interleukin-1α-
00308.2014 dependent senescence-associated secretory phenotype. Arterioscler
114. Ungvari Z, Tarantini S, Hertelendy P, Valcarcel-Ares MN, Fülöp
Thromb Vasc Biol. 2015;35:1963–1974. doi: 10.1161/ATVBAHA.
GA, Logan S, Kiss T, Farkas E, Csiszar A, Yabluchanskiy A. 115.305896
Cerebromicrovascular dysfunction predicts cognitive decline and gait 133. Joaquin AM, Gollapudi S. Functional decline in aging and disease: a role
abnormalities in a mouse model of whole brain irradiation-induced for apoptosis. J Am Geriatr Soc. 2001;49:1234–1240.
accelerated brain senescence. Geroscience. 2017;39:33–42. doi: 134. Asai K, Kudej RK, Shen YT, Yang GP, Takagi G, Kudej AB, Geng YJ,
10.1007/s11357-017-9964-z Sato N, Nazareno JB, Vatner DE, Natividad F, Bishop SP, Vatner SF.
115. Uryga AK, Bennett MR. Ageing induced vascular smooth muscle cell Peripheral vascular endothelial dysfunction and apoptosis in old mon-
senescence in atherosclerosis. J Physiol. 2016;594:2115–2124. doi: keys. Arterioscler Thromb Vasc Biol. 2000;20:1493–1499.
10.1113/JP270923 135. Meng L, Jin W, Wang X. RIP3-mediated necrotic cell death acceler-
116. Durik M, Kavousi M, van der Pluijm I, et al. Nucleotide excision DNA ates systematic inflammation and mortality. Proc Natl Acad Sci USA.
repair is associated with age-related vascular dysfunction. Circulation. 2015;112:11007–11012. doi: 10.1073/pnas.1514730112
2012;126:468–478. 136. Deepa SS, Unnikrishnan A, Matyi S, Hadad N, Richardson A.

117. Matsumoto T, Baker DJ, d’Uscio LV, Mozammel G, Katusic ZS,
Necroptosis increases with age and is reduced by dietary restriction.
van Deursen JM. Aging-associated vascular phenotype in mutant Aging Cell. 2018:e12770.
Ungvari et al   Mechanisms of Vascular Aging   865

137. Wang Q, Liu Z, Ren J, Morgan S, Assa C, Liu B. Receptor-interacting 157. Wang CY, Kim HH, Hiroi Y, Sawada N, Salomone S, Benjamin LE,
protein kinase 3 contributes to abdominal aortic aneurysms via smooth Walsh K, Moskowitz MA, Liao JK. Obesity increases vascular senes-
muscle cell necrosis and inflammation. Circ Res. 2015;116:600–611. cence and susceptibility to ischemic injury through chronic activation of
doi: 10.1161/CIRCRESAHA.116.304899 Akt and mTOR. Sci Signal. 2009;2:ra11. doi: 10.1126/scisignal.2000143
138. Costantino S, Camici GG, Mohammed SA, Volpe M, Lüscher TF, Paneni 158. Yepuri G, Velagapudi S, Xiong Y, Rajapakse AG, Montani JP, Ming XF,
F. Epigenetics and cardiovascular regenerative medicine in the elderly. Yang Z. Positive crosstalk between arginase-II and S6K1 in vascular
Int J Cardiol. 2018;250:207–214. doi: 10.1016/j.ijcard.2017.09.188 endothelial inflammation and aging. Aging Cell. 2012;11:1005–1016.
139. Unnikrishnan A, Jackson J, Matyi SA, Hadad N, Wronowski B,
doi: 10.1111/acel.12001
Georgescu C, Garrett KP, Wren JD, Freeman WM, Richardson A. Role 159. Parlar A, Can C, Erol A, Ulker S. Posttransplantation therapeutic

of DNA methylation in the dietary restriction mediated cellular memory. rapamycin concentration protects nitric oxide-related vascular endo-
Geroscience. 2017;39:331–345. doi: 10.1007/s11357-017-9976-8 thelial function: comparative effects in rat thoracic aorta and coronary
140. Nguyen A, Leblond F, Mamarbachi M, Geoffroy S, Thorin E. Age- endothelial cell culture. Transplant Proc. 2010;42:1923–1930. doi:
dependent demethylation of Sod2 promoter in the mouse femoral artery. 10.1016/j.transproceed.2010.03.134
Oxid Med Cell Longev. 2016;2016:8627384. doi: 10.1155/2016/8627384 160. Cheng C, Tempel D, Oostlander A, Helderman F, Gijsen F, Wentzel J,
141. Connelly JJ, Cherepanova OA, Doss JF, et al. Epigenetic regulation of van Haperen R, Haitsma DB, Serruys PW, van der Steen AF, de Crom R,
COL15A1 in smooth muscle cell replicative aging and atherosclerosis. Krams R. Rapamycin modulates the eNOS vs. shear stress relationship.
Hum Mol Genet. 2013;22:5107–5120. doi: 10.1093/hmg/ddt365 Cardiovasc Res. 2008;78:123–129. doi: 10.1093/cvr/cvm103
142. Toghill BJ, Saratzis A, Harrison SC, Verissimo AR, Mallon EB, Bown 161. Corbin F, Blaise GA, Parent M, Chen H, Daloze PM. Effect of

MJ. The potential role of DNA methylation in the pathogenesis of rapamycin on rat aortic ring vasomotion. J Cardiovasc Pharmacol.
abdominal aortic aneurysm. Atherosclerosis. 2015;241:121–129. doi: 1994;24:813–817.
10.1016/j.atherosclerosis.2015.05.001 162. Milliard S, Silva A, Blaise G, Chen H, Xu D, Qi S, Daloze P. Rapamycin’s
143. Donato AJ, Magerko KA, Lawson BR, Durrant JR, Lesniewski LA, effect on vasomotion in the rat. Transplant Proc. 1998;30:1036–1038.
Seals DR. SIRT-1 and vascular endothelial dysfunction with age- 163. Ha JM, Yun SJ, Kim YW, Jin SY, Lee HS, Song SH, Shin HK, Bae SS.
ing in mice and humans. J Physiol. 2011;589:4545–4554. doi: Platelet-derived growth factor regulates vascular smooth muscle pheno-
10.1113/jphysiol.2011.211219 type via mammalian target of rapamycin complex 1. Biochem Biophys
144. Maeng YS, Kwon JY, Kim EK, Kwon YG. Heterochromatin protein 1 Res Commun. 2015;464:57–62. doi: 10.1016/j.bbrc.2015.05.097
alpha (HP1α: CBX5) is a key regulator in differentiation of endothelial 164. Lesniewski LA, Seals DR, Walker AE, Henson GD, Blimline MW,
progenitor cells to endothelial cells. Stem Cells. 2015;33:1512–1522. Trott DW, Bosshardt GC, LaRocca TJ, Lawson BR, Zigler MC, Donato
doi: 10.1002/stem.1954 AJ. Dietary rapamycin supplementation reverses age-related vascular
145. Suárez Y, Sessa WC. MicroRNAs as novel regulators of angiogenesis. dysfunction and oxidative stress, while modulating nutrient-sensing,
cell cycle, and senescence pathways. Aging Cell. 2017;16:17–26. doi:
Circ Res. 2009;104:442–454. doi: 10.1161/CIRCRESAHA.108.191270
10.1111/acel.12524
146. Feinberg MW, Moore KJ. MicroRNA regulation of atherosclerosis. Circ
165. Chauhan A, Sharma U, Jagannathan NR, Reeta KH, Gupta YK.

Res. 2016;118:703–720. doi: 10.1161/CIRCRESAHA.115.306300
Rapamycin protects against middle cerebral artery occlusion induced
147. Gareri C, De Rosa S, Indolfi C. MicroRNAs for restenosis and throm-
focal cerebral ischemia in rats. Behav Brain Res. 2011;225:603–609. doi:
bosis after vascular injury. Circ Res. 2016;118:1170–1184. doi:
10.1016/j.bbr.2011.08.035
10.1161/CIRCRESAHA.115.308237
166. Zhang W, Khatibi NH, Yamaguchi-Okada M, Yan J, Chen C, Hu

148. Ungvari Z, Tucsek Z, Sosnowska D, Toth P, Gautam T, Podlutsky A,
Downloaded from http://ahajournals.org by on June 28, 2023

Q, Meng H, Han H, Liu S, Zhou C. Mammalian target of rapamycin


Csiszar A, Losonczy G, Valcarcel-Ares MN, Sonntag WE, Csiszar A.
(mTOR) inhibition reduces cerebral vasospasm following a subarach-
Aging-induced dysregulation of dicer1-dependent microRNA expres-
noid hemorrhage injury in canines. Exp Neurol. 2012;233:799–806. doi:
sion impairs angiogenic capacity of rat cerebromicrovascular endo-
10.1016/j.expneurol.2011.11.046
thelial cells. J Gerontol A Biol Sci Med Sci. 2013;68:877–891. doi:
167. Fletcher L, Evans TM, Watts LT, Jimenez DF, Digicaylioglu M.

10.1093/gerona/gls242
Rapamycin treatment improves neuron viability in an in vitro model of
149. Menghini R, Stöhr R, Federici M. MicroRNAs in vascular aging and
stroke. PLoS One. 2013;8:e68281. doi: 10.1371/journal.pone.0068281
atherosclerosis. Ageing Res Rev. 2014;17:68–78. doi: 10.1016/j.arr. 168. Yin L, Ye S, Chen Z, Zeng Y. Rapamycin preconditioning attenuates tran-
2014.03.005 sient focal cerebral ischemia/reperfusion injury in mice. Int J Neurosci.
150. Tarantini S, Giles CB, Wren JD, Ashpole NM, Valcarcel-Ares MN, Wei 2012;122:748–756. doi: 10.3109/00207454.2012.721827
JY, Sonntag WE, Ungvari Z, Csiszar A. IGF-1 deficiency in a critical 169. Spilman P, Podlutskaya N, Hart MJ, Debnath J, Gorostiza O, Bredesen
period early in life influences the vascular aging phenotype in mice by D, Richardson A, Strong R, Galvan V. Inhibition of mTOR by rapamy-
altering miRNA-mediated post-transcriptional gene regulation: implica- cin abolishes cognitive deficits and reduces amyloid-beta levels in a
tions for the developmental origins of health and disease hypothesis. Age mouse model of Alzheimer’s disease. PLoS One. 2010;5:e9979. doi:
(Dordr). 2016;38:239–258. doi: 10.1007/s11357-016-9943-9 10.1371/journal.pone.0009979
151. Gupta SK, Piccoli MT, Thum T. Non-coding RNAs in cardiovascular 170. Lin AL, Jahrling JB, Zhang W, DeRosa N, Bakshi V, Romero P, Galvan
ageing. Ageing Res Rev. 2014;17:79–85. doi: 10.1016/j.arr.2014.01.002 V, Richardson A. Rapamycin rescues vascular, metabolic and learning
152. Boon RA, Hofmann P, Michalik KM, Lozano-Vidal N, Berghäuser D, deficits in apolipoprotein E4 transgenic mice with pre-symptomatic
Fischer A, Knau A, Jaé N, Schürmann C, Dimmeler S. Long noncoding Alzheimer’s disease. J Cereb Blood Flow Metab. 2017;37:217–226. doi:
RNA Meg3 controls endothelial cell aging and function: implications for 10.1177/0271678X15621575
regenerative angiogenesis. J Am Coll Cardiol. 2016;68:2589–2591. doi: 171. Van Skike CE, Jahrling JB, Olson AB, Sayre NL, Hussong SA, Ungvari
10.1016/j.jacc.2016.09.949 Z, Lechleiter JD, Galvan V. Inhibition of mTOR protects the blood-brain
153. Sabatini DM. Twenty-five years of mTOR: uncovering the link from barrier in models of Alzheimer’s disease and vascular cognitive impair-
nutrients to growth. Proc Natl Acad Sci USA. 2017;114:11818–11825. ment. Am J Physiol Heart Circ Physiol. 2018;314:H693–H703. doi:
doi: 10.1073/pnas.1716173114 10.1152/ajpheart.00570.2017
154. Johnson SC, Sangesland M, Kaeberlein M, Rabinovitch PS. Modulating 172. Van Skike CE, Galvan V. A perfect sTORm: the role of the mamma-
mTOR in aging and health. Interdiscip Top Gerontol. 2015;40:107–127. lian target of rapamycin (mTOR) in cerebrovascular dysfunction of
doi: 10.1159/000364974 Alzheimer’s disease: a mini-review. Gerontology. 2018;64:205–211. doi:
155. Caccamo A, Majumder S, Richardson A, Strong R, Oddo S. Molecular 10.1159/000485381
interplay between mammalian target of rapamycin (mTOR), amy- 173. Jahrling JB, Lin AL, DeRosa N, Hussong SA, Van Skike CE, Girotti M,
loid-beta, and Tau: effects on cognitive impairments. J Biol Chem. Javors M, Zhao Q, Maslin LA, Asmis R, Galvan V. mTOR drives cere-
2010;285:13107–13120. doi: 10.1074/jbc.M110.100420 bral blood flow and memory deficits in LDLR-/- mice modeling athero-
156. Lin AL, Zheng W, Halloran JJ, Burbank RR, Hussong SA, Hart MJ, sclerosis and vascular cognitive impairment. J Cereb Blood Flow Metab.
Javors M, Shih YY, Muir E, Solano Fonseca R, Strong R, Richardson AG, 2018;38:58–74. doi: 10.1177/0271678X17705973
Lechleiter JD, Fox PT, Galvan V. Chronic rapamycin restores brain vas- 174. Fry JL, Al Sayah L, Weisbrod RM, Van Roy I, Weng X, Cohen RA,
cular integrity and function through NO synthase activation and improves Bachschmid MM, Seta F. Vascular smooth muscle sirtuin-1 protects
memory in symptomatic mice modeling Alzheimer’s disease. J Cereb against diet-induced aortic stiffness. Hypertension. 2016;68:775–784.
Blood Flow Metab. 2013;33:1412–1421. doi: 10.1038/jcbfm.2013.82 doi: 10.1161/HYPERTENSIONAHA.116.07622
866  Circulation Research  September 14, 2018

175. Chen YX, Zhang M, Cai Y, Zhao Q, Dai W. The Sirt1 activator SRT1720 195. Basso N, Cini R, Pietrelli A, Ferder L, Terragno NA, Inserra F. Protective
attenuates angiotensin II-induced atherosclerosis in apoE⁻/⁻ mice through effect of long-term angiotensin II inhibition. Am J Physiol Heart Circ
inhibiting vascular inflammatory response. Biochem Biophys Res Physiol. 2007;293:H1351–H1358. doi: 10.1152/ajpheart.00393.2007
Commun. 2015;465:732–738. doi: 10.1016/j.bbrc.2015.08.066 196. de Cavanagh EM, Inserra F, Ferder L. Angiotensin II blockade: how its
176. Mattison JA, Wang M, Bernier M, et al. Resveratrol prevents high
molecular targets may signal to mitochondria and slow aging. Coincidences
fat/sucrose diet-induced central arterial wall inflammation and stiff- with calorie restriction and mTOR inhibition. Am J Physiol Heart Circ
ening in nonhuman primates. Cell Metab. 2014;20:183–190. doi: Physiol. 2015;309:H15–H44. doi: 10.1152/ajpheart.00459.2014
10.1016/j.cmet.2014.04.018 197. Benigni A, Corna D, Zoja C, Sonzogni A, Latini R, Salio M, Conti S,
177. Baur JA, Ungvari Z, Minor RK, Le Couteur DG, de Cabo R. Are sirtu- Rottoli D, Longaretti L, Cassis P, Morigi M, Coffman TM, Remuzzi G.
ins viable targets for improving healthspan and lifespan? Nat Rev Drug Disruption of the Ang II type 1 receptor promotes longevity in mice. J
Discov. 2012;11:443–461. doi: 10.1038/nrd3738 Clin Invest. 2009;119:524–530. doi: 10.1172/JCI36703
178. Liu Y, Wang TT, Zhang R, Fu WY, Wang X, Wang F, Gao P, Ding YN, Xie 198. Spinetti G, Wang M, Monticone R, Zhang J, Zhao D, Lakatta EG. Rat
Y, Hao DL, Chen HZ, Liu DP. Calorie restriction protects against experi- aortic MCP-1 and its receptor CCR2 increase with age and alter vas-
mental abdominal aortic aneurysms in mice. J Exp Med. 2016;213:2473– cular smooth muscle cell function. Arterioscler Thromb Vasc Biol.
2488. doi: 10.1084/jem.20151794 2004;24:1397–1402. doi: 10.1161/01.ATV.0000134529.65173.08
179. Cardus A, Uryga AK, Walters G, Erusalimsky JD. SIRT6 protects human 199. Wang M, Takagi G, Asai K, Resuello RG, Natividad FF, Vatner DE,
endothelial cells from DNA damage, telomere dysfunction, and senes- Vatner SF, Lakatta EG. Aging increases aortic MMP-2 activity and angio-
cence. Cardiovasc Res. 2013;97:571–579. doi: 10.1093/cvr/cvs352 tensin II in nonhuman primates. Hypertension. 2003;41:1308–1316. doi:
180. Xu S, Yin M, Koroleva M, Mastrangelo MA, Zhang W, Bai P, Little 10.1161/01.HYP.0000073843.56046.45
PJ, Jin ZG. SIRT6 protects against endothelial dysfunction and ath- 200. Wang M, Zhang J, Spinetti G, Jiang LQ, Monticone R, Zhao D, Cheng
erosclerosis in mice. Aging (Albany NY). 2016;8:1064–1082. doi: L, Krawczyk M, Talan M, Pintus G, Lakatta EG. Angiotensin II activates
10.18632/aging.100975 matrix metalloproteinase type II and mimics age-associated carotid arte-
181. Gorenne I, Kumar S, Gray K, Figg N, Yu H, Mercer J, Bennett M. rial remodeling in young rats. Am J Pathol. 2005;167:1429–1442. doi:
Vascular smooth muscle cell sirtuin 1 protects against DNA dam- 10.1016/S0002-9440(10)61229-1
age and inhibits atherosclerosis. Circulation. 2013;127:386–396. doi: 201. Hayashi K, Miyagawa K, Sato K, Ueda R, Dohi Y. Temocapril, an angio-
10.1161/CIRCULATIONAHA.112.124404 tensin converting enzyme inhibitor, ameliorates age-related increase
182. Das DK, Maulik N. Resveratrol in cardioprotection: a therapeutic promise in carotid arterial stiffness in normotensive subjects. Cardiology.
of alternative medicine. Mol Interv. 2006;6:36–47. doi: 10.1124/mi.6.1.7 2006;106:190–194. doi: 10.1159/000093024
183. Stivala LA, Savio M, Carafoli F, Perucca P, Bianchi L, Maga G, Forti 202. Michel JB, Heudes D, Michel O, Poitevin P, Philippe M, Scalbert E,
L, Pagnoni UM, Albini A, Prosperi E, Vannini V. Specific structural Corman B, Levy BI. Effect of chronic ANG I-converting enzyme inhibi-
determinants are responsible for the antioxidant activity and the cell tion on aging processes. II. Large arteries. Am J Physiol. 1994;267:R124–
cycle effects of resveratrol. J Biol Chem. 2001;276:22586–22594. doi: R135. doi: 10.1152/ajpregu.1994.267.1.R124
10.1074/jbc.M101846200 203. Dai DF, Rabinovitch PS, Ungvari Z. Mitochondria and cardiovascular
184. Yoshino J, Mills KF, Yoon MJ, Imai S. Nicotinamide mononucleotide, aging. Circ Res. 2012;110:1109–1124. doi: 10.1161/CIRCRESAHA.
a key NAD(+) intermediate, treats the pathophysiology of diet- and 111.246140
age-induced diabetes in mice. Cell Metab. 2011;14:528–536. doi: 204. Toth P, Tucsek Z, Sosnowska D, Gautam T, Mitschelen M, Tarantini
10.1016/j.cmet.2011.08.014 S, Deak F, Koller A, Sonntag WE, Csiszar A, Ungvari Z. Age-related
Downloaded from http://ahajournals.org by on June 28, 2023

185. Hattori Y, Suzuki K, Hattori S, Kasai K. Metformin inhibits cytokine- autoregulatory dysfunction and cerebromicrovascular injury in mice
induced nuclear factor kappaB activation via AMP-activated pro- with angiotensin II-induced hypertension. J Cereb Blood Flow Metab.
tein kinase activation in vascular endothelial cells. Hypertension. 2013;33:1732–1742. doi: 10.1038/jcbfm.2013.143
2006;47:1183–1188. doi: 10.1161/01.HYP.0000221429.94591.72 205. Krug AW, Allenhöfer L, Monticone R, Spinetti G, Gekle M, Wang M,
186. Hardie DG, Hawley SA, Scott JW. AMP-activated protein kinase–devel- Lakatta EG. Elevated mineralocorticoid receptor activity in aged rat vas-
opment of the energy sensor concept. J Physiol. 2006;574:7–15. doi: cular smooth muscle cells promotes a proinflammatory phenotype via
10.1113/jphysiol.2006.108944 extracellular signal-regulated kinase ½ mitogen-activated protein kinase
187. Barzilai N, Crandall JP, Kritchevsky SB, Espeland MA. Metformin
and epidermal growth factor receptor-dependent pathways. Hypertension.
as a tool to target aging. Cell Metab. 2016;23:1060–1065. doi: 2010;55:1476–1483. doi: 10.1161/HYPERTENSIONAHA.109.148783
10.1016/j.cmet.2016.05.011 206. Ewald CY, Landis JN, Porter Abate J, Murphy CT, Blackwell TK. Dauer-
188. Chen ZP, Mitchelhill KI, Michell BJ, Stapleton D, Rodriguez-Crespo independent insulin/IGF-1-signalling implicates collagen remodelling in
I, Witters LA, Power DA, Ortiz de Montellano PR, Kemp BE. AMP- longevity. Nature. 2015;519:97–101. doi: 10.1038/nature14021
activated protein kinase phosphorylation of endothelial NO synthase. 207. Phillip JM, Aifuwa I, Walston J, Wirtz D. The mechanobiology of
FEBS Lett. 1999;443:285–289. aging. Annu Rev Biomed Eng. 2015;17:113–141. doi: 10.1146/
189. Chen Z, Peng IC, Sun W, Su MI, Hsu PH, Fu Y, Zhu Y, DeFea K, Pan S, annurev-bioeng-071114-040829
Tsai MD, Shyy JY. AMP-activated protein kinase functionally phosphor- 208. Jacob MP. Extracellular matrix remodeling and matrix metalloprotein-
ylates endothelial nitric oxide synthase Ser633. Circ Res. 2009;104:496– ases in the vascular wall during aging and in pathological conditions.
505. doi: 10.1161/CIRCRESAHA.108.187567 Biomed Pharmacother. 2003;57:195–202.
190. Levine YC, Li GK, Michel T. Agonist-modulated regulation of AMP- 209. Tarantini S, Valcarcel-Ares NM, Yabluchanskiy A, Springo Z, Fulop
activated protein kinase (AMPK) in endothelial cells. Evidence for an GA, Ashpole N, Gautam T, Giles CB, Wren JD, Sonntag WE, Csiszar A,
AMPK -> Rac1 -> Akt -> endothelial nitric-oxide synthase pathway. J Ungvari Z. Insulin-like growth factor 1 deficiency exacerbates hyperten-
Biol Chem. 2007;282:20351–20364. doi: 10.1074/jbc.M702182200 sion-induced cerebral microhemorrhages in mice, mimicking the aging
191. Nagata D, Mogi M, Walsh K. AMP-activated protein kinase (AMPK) phenotype. Aging Cell. 2017;16:469–479. doi: 10.1111/acel.12583
signaling in endothelial cells is essential for angiogenesis in response 210. Dao HH, Essalihi R, Bouvet C, Moreau P. Evolution and modulation of
to hypoxic stress. J Biol Chem. 2003;278:31000–31006. doi: age-related medial elastocalcinosis: impact on large artery stiffness and
10.1074/jbc.M300643200 isolated systolic hypertension. Cardiovasc Res. 2005;66:307–317. doi:
192. Lesniewski LA, Zigler MC, Durrant JR, Donato AJ, Seals DR. Sustained 10.1016/j.cardiores.2005.01.012
activation of AMPK ameliorates age-associated vascular endothelial dys- 211. Cantini C, Kieffer P, Corman B, Limiñana P, Atkinson J, Lartaud-

function via a nitric oxide-independent mechanism. Mech Ageing Dev. Idjouadiene I. Aminoguanidine and aortic wall mechanics, structure, and
2012;133:368–371. doi: 10.1016/j.mad.2012.03.011 composition in aged rats. Hypertension. 2001;38:943–948.
193. Pu Y, Zhang H, Wang P, Zhao Y, Li Q, Wei X, Cui Y, Sun J, Shang 212. Scuteri A, Morrell CH, Orrù M, Strait JB, Tarasov KV, Ferreli LA, Loi
Q, Liu D, Zhu Z. Dietary curcumin ameliorates aging-related cere- F, Pilia MG, Delitala A, Spurgeon H, Najjar SS, AlGhatrif M, Lakatta
brovascular dysfunction through the AMPK/uncoupling protein 2 EG. Longitudinal perspective on the conundrum of central arterial stiff-
pathway. Cell Physiol Biochem. 2013;32:1167–1177. doi: 10.1159/ ness, blood pressure, and aging. Hypertension. 2014;64:1219–1227. doi:
000354516 10.1161/HYPERTENSIONAHA.114.04127
194. Kumar S, Dietrich N, Kornfeld K. Angiotensin converting enzyme
213. Rosenberg GA. Extracellular matrix inflammation in vascular cogni-
(ACE) inhibitor extends caenorhabditis elegans life span. PLoS Genet. tive impairment and dementia. Clin Sci (Lond). 2017;131:425–437. doi:
2016;12:e1005866. doi: 10.1371/journal.pgen.1005866 10.1042/CS20160604
Ungvari et al   Mechanisms of Vascular Aging   867

214. Pascual G, Mendieta C, García-Honduvilla N, Corrales C, Bellón JM, carotid intimal thickening. Basic Res Cardiol. 2008;103:582–586. doi:
Buján J. TGF-beta1 upregulation in the aging varicose vein. J Vasc Res. 10.1007/s00395-008-0742-z
2007;44:192–201. doi: 10.1159/000100375 233. Heiss C, Keymel S, Niesler U, Ziemann J, Kelm M, Kalka C. Impaired
215. Khan AS, Lynch CD, Sane DC, Willingham MC, Sonntag WE. Growth progenitor cell activity in age-related endothelial dysfunction. J Am Coll
hormone increases regional coronary blood flow and capillary density in Cardiol. 2005;45:1441–1448. doi: 10.1016/j.jacc.2004.12.074
aged rats. J Gerontol A Biol Sci Med Sci. 2001;56:B364–B371. 234. Rauscher FM, Goldschmidt-Clermont PJ, Davis BH, Wang T, Gregg D,
216. Tarantini S, Tucsek Z, Valcarcel-Ares MN, Toth P, Gautam T, Giles Ramaswami P, Pippen AM, Annex BH, Dong C, Taylor DA. Aging, pro-
CB, Ballabh P, Wei JY, Wren JD, Ashpole NM, Sonntag WE, Ungvari genitor cell exhaustion, and atherosclerosis. Circulation. 2003;108:457–
Z, Csiszar A. Circulating IGF-1 deficiency exacerbates hypertension- 463. doi: 10.1161/01.CIR.0000082924.75945.48
induced microvascular rarefaction in the mouse hippocampus and retro- 235. Zhu G, Song M, Wang H, Zhao G, Yu Z, Yin Y, Zhao X, Huang L.
splenial cortex: implications for cerebromicrovascular and brain aging. Young environment reverses the declined activity of aged rat-derived
Age (Dordr). 2016;38:273–289. doi: 10.1007/s11357-016-9931-0 endothelial progenitor cells: involvement of the phosphatidylinositol
217. Toth P, Tarantini S, Ashpole NM, Tucsek Z, Milne GL, Valcarcel-
3-kinase/Akt signaling pathway. Ann Vasc Surg. 2009;23:519–534. doi:
Ares NM, Menyhart A, Farkas E, Sonntag WE, Csiszar A, Ungvari Z. 10.1016/j.avsg.2008.11.013
IGF-1 deficiency impairs neurovascular coupling in mice: implications 236. Hayek SS, MacNamara J, Tahhan AS, et al. Circulating progenitor

for cerebromicrovascular aging. Aging Cell. 2015;14:1034–1044. doi: cells identify peripheral arterial disease in patients with coronary artery
10.1111/acel.12372 disease. Circ Res. 2016;119:564–571. doi: 10.1161/CIRCRESAHA.
218. Toth P, Tucsek Z, Tarantini S, Sosnowska D, Gautam T, Mitschelen M, 116.308802
Koller A, Sonntag WE, Csiszar A, Ungvari Z. IGF-1 deficiency impairs 237. Mekonnen G, Hayek SS, Mehta PK, et al. Circulating progenitor cells
cerebral myogenic autoregulation in hypertensive mice. J Cereb Blood and coronary microvascular dysfunction: results from the NHLBI-
Flow Metab. 2014;34:1887–1897. doi: 10.1038/jcbfm.2014.156 sponsored Women’s Ischemia Syndrome Evaluation - Coronary Vascular
219. Higashi Y, Sukhanov S, Anwar A, Shai SY, Delafontaine P. Aging, ath- Dysfunction Study (WISE-CVD). Atherosclerosis. 2016;253:111–117.
erosclerosis, and IGF-1. J Gerontol A Biol Sci Med Sci. 2012;67:626– doi: 10.1016/j.atherosclerosis.2016.08.026
639. doi: 10.1093/gerona/gls102 238. Dawson J, Tooze J, Cockerill G, Choke E, Loftus I, Thompson MM.
220. Lähteenvuo J, Rosenzweig A. Effects of aging on angiogenesis. Circ Res. Endothelial progenitor cells and abdominal aortic aneurysms. Ann NY
2012;110:1252–1264. doi: 10.1161/CIRCRESAHA.111.246116 Acad Sci. 2006;1085:327–330. doi: 10.1196/annals.1383.011
221. Katsimpardi L, Litterman NK, Schein PA, Miller CM, Loffredo FS, 239. Vaughan KL, Kaiser T, Peaden R, Anson RM, de Cabo R, Mattison JA.
Wojtkiewicz GR, Chen JW, Lee RT, Wagers AJ, Rubin LL. Vascular and Caloric restriction study design limitations in rodent and nonhuman
neurogenic rejuvenation of the aging mouse brain by young systemic primate studies. J Gerontol A Biol Sci Med Sci. 2017;73:48–53. doi:
factors. Science. 2014;344:630–634. doi: 10.1126/science.1251141 10.1093/gerona/glx088
222. Ingraham JP, Forbes ME, Riddle DR, Sonntag WE. Aging reduces
240. Donato AJ, Walker AE, Magerko KA, Bramwell RC, Black AD, Henson
hypoxia-induced microvascular growth in the rodent hippocampus. J GD, Lawson BR, Lesniewski LA, Seals DR. Life-long caloric restric-
Gerontol A Biol Sci Med Sci. 2008;63:12–20. tion reduces oxidative stress and preserves nitric oxide bioavailability
223. Ungvari Z, Parrado-Fernandez C, Csiszar A, de Cabo R. Mechanisms and function in arteries of old mice. Aging Cell. 2013;12:772–783. doi:
underlying caloric restriction and lifespan regulation: implications 10.1111/acel.12103
for vascular aging. Circ Res. 2008;102:519–528. doi: 10.1161/ 241. Edwards AG, Donato AJ, Lesniewski LA, Gioscia RA, Seals DR, Moore
CIRCRESAHA.107.168369 RL. Life-long caloric restriction elicits pronounced protection of the
Downloaded from http://ahajournals.org by on June 28, 2023

224. Nisoli E, Tonello C, Cardile A, Cozzi V, Bracale R, Tedesco L, Falcone aged myocardium: a role for AMPK. Mech Ageing Dev. 2010;131:739–
S, Valerio A, Cantoni O, Clementi E, Moncada S, Carruba MO. Calorie 742. doi: 10.1016/j.mad.2010.09.007
restriction promotes mitochondrial biogenesis by inducing the expression 242. Colman RJ, Beasley TM, Kemnitz JW, Johnson SC, Weindruch

of eNOS. Science. 2005;310:314–317. doi: 10.1126/science.1117728 R, Anderson RM. Caloric restriction reduces age-related and all-
225. Kondo M, Shibata R, Miura R, Shimano M, Kondo K, Li P, Ohashi T, cause mortality in rhesus monkeys. Nat Commun. 2014;5:3557. doi:
Kihara S, Maeda N, Walsh K, Ouchi N, Murohara T. Caloric restriction 10.1038/ncomms4557
stimulates revascularization in response to ischemia via adiponectin- 243. Cefalu WT, Wang ZQ, Bell-Farrow AD, Collins J, Morgan T, Wagner
mediated activation of endothelial nitric-oxide synthase. J Biol Chem. JD. Caloric restriction and cardiovascular aging in cynomolgus monkeys
2009;284:1718–1724. doi: 10.1074/jbc.M805301200 (Macaca fascicularis): metabolic, physiologic, and atherosclerotic mea-
226. Pierce GL, Beske SD, Lawson BR, Southall KL, Benay FJ, Donato AJ, sures from a 4-year intervention trial. J Gerontol A Biol Sci Med Sci.
Seals DR. Weight loss alone improves conduit and resistance artery endo- 2004;59:1007–1014.
thelial function in young and older overweight/obese adults. Hypertension. 244. Wycherley TP, Brinkworth GD, Noakes M, Buckley JD, Clifton

2008;52:72–79. doi: 10.1161/HYPERTENSIONAHA.108.111427 PM. Effect of caloric restriction with and without exercise train-
227. Csiszar A, Sosnowska D, Tucsek Z, Gautam T, Toth P, Losonczy
ing on oxidative stress and endothelial function in obese subjects
G, Colman RJ, Weindruch R, Anderson RM, Sonntag WE, Ungvari with type 2 diabetes. Diabetes Obes Metab. 2008;10:1062–1073. doi:
Z. Circulating factors induced by caloric restriction in the nonhu- 10.1111/j.1463-1326.2008.00863.x
man primate Macaca mulatta activate angiogenic processes in endo- 245. Csiszar A, Labinskyy N, Zhao X, Hu F, Serpillon S, Huang Z,

thelial cells. J Gerontol A Biol Sci Med Sci. 2013;68:235–249. doi: Ballabh P, Levy RJ, Hintze TH, Wolin MS, Austad SN, Podlutsky
10.1093/gerona/gls158 A, Ungvari Z. Vascular superoxide and hydrogen peroxide produc-
228. Allard JS, Heilbronn LK, Smith C, Hunt ND, Ingram DK, Ravussin E, tion and oxidative stress resistance in two closely related rodent spe-
de Cabo R; Pennington CALERIE Team. In vitro cellular adaptations cies with disparate longevity. Aging Cell. 2007;6:783–797. doi:
of indicators of longevity in response to treatment with serum collected 10.1111/j.1474-9726.2007.00339.x
from humans on calorie restricted diets. PLoS One. 2008;3:e3211. doi: 246. Labinskyy N, Mukhopadhyay P, Toth J, Szalai G, Veres M, Losonczy
10.1371/journal.pone.0003211 G, Pinto JT, Pacher P, Ballabh P, Podlutsky A, Austad SN, Csiszar A,
229. Shinmura K, Tamaki K, Bolli R. Impact of 6-mo caloric restriction on Ungvari Z. Longevity is associated with increased vascular resis-
myocardial ischemic tolerance: possible involvement of nitric oxide- tance to high glucose-induced oxidative stress and inflammatory gene
dependent increase in nuclear Sirt1. Am J Physiol Heart Circ Physiol. expression in Peromyscus leucopus. Am J Physiol Heart Circ Physiol.
2008;295:H2348–H2355. doi: 10.1152/ajpheart.00602.2008 2009;296:H946–H956. doi: 10.1152/ajpheart.00693.2008
230. Shinmura K, Tamaki K, Saito K, Nakano Y, Tobe T, Bolli R.
247. Ungvari Z, Krasnikov BF, Csiszar A, Labinskyy N, Mukhopadhyay P,
Cardioprotective effects of short-term caloric restriction are mediated by Pacher P, Cooper AJ, Podlutskaya N, Austad SN, Podlutsky A. Testing
adiponectin via activation of AMP-activated protein kinase. Circulation. hypotheses of aging in long-lived mice of the genus peromyscus: asso-
2007;116:2809–2817. doi: 10.1161/CIRCULATIONAHA.107.725697 ciation between longevity and mitochondrial stress resistance, ROS
231. Niemann B, Silber RE, Rohrbach S. Age-specific effects of short- and detoxification pathways, and DNA repair efficiency. Age (Dordr).
long-term caloric restriction on the expression of adiponectin and adipo- 2008;30:121–133. doi: 10.1007/s11357-008-9059-y
nectin receptors: influence of intensity of food restriction. Exp Gerontol. 248. Kress BT, Iliff JJ, Xia M, Wang M, Wei HS, Zeppenfeld D, Xie L,
2008;43:706–713. doi: 10.1016/j.exger.2008.02.008 Kang H, Xu Q, Liew JA, Plog BA, Ding F, Deane R, Nedergaard M.
232. Keymel S, Kalka C, Rassaf T, Yeghiazarians Y, Kelm M, Heiss C.
Impairment of paravascular clearance pathways in the aging brain. Ann
Impaired endothelial progenitor cell function predicts age-dependent Neurol. 2014;76:845–861. doi: 10.1002/ana.24271

You might also like