Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2016 Dec 1.
Published in final edited form as: J Mol Cell Cardiol. 2015 Feb 2;89(0 0):122–135. doi: 10.1016/j.yjmcc.2015.01.021

Cellular and Molecular Biology of Aging Endothelial Cells

Anthony J Donato 1,2, R Garrett Morgan 3, Ashley E Walker 1, Lisa A Lesniewski 1,2
PMCID: PMC4522407  NIHMSID: NIHMS665826  PMID: 25655936

Abstract

Cardiovascular disease (CVD) is the leading cause of death in the United States and aging is a major risk factor for CVD development. One of the major age-related arterial phenotypes thought to be responsible for the development of CVD in older adults is endothelial dysfunction. Endothelial function is modulated by traditional CVD risk factors in young adults, but advancing age is independently associated with the development of vascular endothelial dysfunction. This endothelial dysfunction results from a reduction in nitric oxide bioavailability downstream of endothelial oxidative stress and inflammation that can be further modulated by traditional CVD risk factors in older adults. Greater endothelial oxidative stress with aging is a result of augmented production from the intracellular enzymes NADPH oxidase and uncoupled eNOS, as well as from mitochondrial respiration in the absence of appropriate increases in antioxidant defenses as regulated by relevant transcription factors, such as FOXO. Interestingly, it appears that NFkB, a critical inflammatory transcription factor, is sensitive to this age-related endothelial redox change and its activation induces transcription of pro-inflammatory cytokines that can further suppress endothelial function, thus creating a vicious feed-forward cycle. This review will discuss the two macro-mechanistic processes, oxidative stress and inflammation, that contribute to endothelial dysfunction with advancing age as well as the cellular and molecular events that lead to the vicious cycle of inflammation and oxidative stress in the aged endothelium. Other potential mediators of this pro-inflammatory endothelial phenotype are increases in immune or senescent cells in the vasculature. Of note, genomic instability, telomere dysfunction or DNA damage have been shown to trigger cell senescence via the p53/p21 pathway that results in increased inflammatory signaling in arteries from older adults. This review will discuss the current state of knowledge regarding the emerging concepts of senescence and genomic instability as mechanisms underlying oxidative stress and inflammation in the aged endothelium. Lastly, energy sensitive/stress resistance pathways (SIRT-1, AMPK, mTOR) are altered in endothelial cells and/or arteries with aging and these pathways may modulate endothelial function via key oxidative stress and inflammation-related transcription factors. This review will also discuss what is known about the role of “energy sensing” longevity pathways in modulating endothelial function with advancing age. With the growing population of older adults, elucidating the cellular and molecular mechanisms of endothelial dysfunction with age is critical to establishing appropriate and measured strategies to utilize pharmacological and lifestyle interventions aimed at alleviating CVD risk.

Keywords: Aging, Endothelium, Oxidative Stress, Inflammation, Senescence, Telomere, Genomic Instability, SIRT-1, AMPK, mTOR

1. Cardiovascular Disease, Vascular Endothelial Function, and Aging

Cardiovascular diseases (CVD), largely defined as stroke, coronary artery disease, heart failure, and cardiac arrest, are the predominant killers of Americans, accounting for ~752,000 deaths per year according to current statistics from the Centers for Disease Control [1]. CVDs cause ~35% of all deaths for Americans 65 years of age or older, making them the leading causes of death in this age group [1]. Furthermore, with advancing age, the prevalence of CVDs among Americans increases progressively, from ~5.5% in 25–44 year olds to ~41% in people 65 years of age or older [1]. Thus, CVDs can be considered true diseases of aging.

Heart disease, stroke, and hypertension are all diseases currently recognized to be caused, in part, by arterial dysfunction [2, 3]. Age-related alterations to arteries are thought to lead to a dysfunctional phenotype that precedes CVDs [2, 4, 5]. Importantly, the dysfunctional phenotype that develops in arteries with advancing age can occur in the absence of overt CVD and conventional CVD risk factors [6, 7], supporting the idea that these changes are a primary effect of aging that may be a precursor to the development of CVD. Large landmark studies, like the Baltimore Longitudinal Study on Aging (BLSA) and the Framingham Heart Study, demonstrated that the age-associated phenotype of arteries involves, among other changes, the development of a dysfunctional arterial endothelium [4, 810]. This dysfunctional endothelial phenotype is common to humans and non-human primates as well as rodents [1113] and these changes contribute to a host of hemodynamic changes, including augmented large and resistance arterial tone, induction of greater oscillatory shear stress and elevated large artery stiffness[2, 10], that contribute to increases in arterial blood pressure [14] and atherosclerosis [3] seen with advancing age.

The arterial endothelium is extremely dynamic and performs many vital functions that vary from one segment of the arterial tree to another as well as from one organ system to another [15, 16]. The vascular endothelium releases molecules that act in an autocrine and paracrine manner to regulate the function and health of the vascular network. These include the maintenance of blood in a fluid state; exchange of fluid and molecules between the blood and surrounding tissues; creation of new vascular networks; participation and facilitation of the immune response; and the control of vascular resistance in response to changes in blood flow by regulating arterial tone in resistance arteries [15, 16]. A healthy vascular endothelium is in a tightly regulated state of balance between pro- and anti-oxidants, vasodilators and vasoconstrictors, pro- and anti-inflammatory molecules, and pro- and anti-thrombotic signals. A diseased or dysfunctional endothelium that has lost its tightly regulated balance and displays pro-oxidant, vasoconstrictor, pro-inflammatory and pro-thrombotic properties. One hallmark of vascular endothelial dysfunction is impaired endothelial dependent dilation (EDD), which is predictive of future CVD events [3, 17]. Indeed, the Framingham Heart Study has recently demonstrated that increased age is the strongest independent correlate of EDD [18]. Therefore, it is of great clinical significance that we obtain a better understanding of the mechanisms underlying age-related decreases in endothelial function and to test the efficacy of interventions that may restore endothelial function in middle-aged and older adults.

2. Goal of the Review

The first goal of this review will be to introduce the two macro-mechanistic processes, oxidative stress and inflammation, that contribute to endothelial dysfunction in healthy older adults and rodent models. Next, we will discuss the cellular and molecular events that lead to the vicious cycle of inflammation and oxidative stress in the aged endothelium. Then, we will discuss the emerging concepts of senescence and genomic instability as it relates to the aforementioned processes. Lastly, we explore how “energy sensing” longevity pathways that appear to modulate endothelial function with advancing age. In this review, we will focus first on in vivo or ex vivo studies that directly examined endothelial cells. However, a major obstacle to our understanding of the events that lead to endothelial dysfunction with advancing age is access to pure primary endothelial cells from humans or rodent models. Next, we will consider studies utilizing whole artery homogenates with the understanding that protein expression in whole arteries is strongly biased toward the smooth muscle cell component rather than the endothelium. Lastly, this review will explore the mechanisms of endothelium dysfunction defined as reductions in EDD assessed by the response to pharmacological or physiological stimuli. Our focus on EDD is because a majority of studies that perform mechanistic studies utilize this marker. It is not to say measures of angiogenesis, permeability, fibrinolysis or other markers of endothelial function are not as important; indeed these are critical functions which are vastly understudied, but at the present time, the mechanisms leading to impairments in these functions in aged endothelial cells or their direct relation to CVD development are not clearly understood.

3. Mechanisms of Age-Associated Vascular Endothelial Dysfunction

Aging is associated with endothelial dysfunction in both men and women in the absence of clinical disease. A majority of the published studies describing this dysfunction measure EDD [1924], but measures of fibrinolytic factors released from the endothelium [25], permeability [26] and angiogenesis [27, 28] have also been made. Multiple animal models of vascular aging phenocopy findings from human studies, have demonstrated impaired EDD [29], fibrinolysis [30], permeability [31], and angiogenesis [32] with advancing age. In older adults, several traditional fasted risk factors, i.e., elevated blood pressure [33], LDL cholesterol [34], blood glucose [35], sodium intake [36], as well as non-traditional markers, i.e., white blood cell count [37] and plasma norepinephrine [38], can modulate the severity of endothelial dysfunction as assessed by EDD (as reviewed in depth by Seals et al.[6]). Interestingly most of these factors have been shown to modulate endothelial function via oxidative stress or inflammation, as described in detail below. Lastly, it is known that with advancing age post prandial clearance of elevated glucose and lipids decreases significantly even in healthy adults [39, 40]. Furthermore, this post prandial state is associated with acute endothelial dysfunction in young and middle aged adults [41, 42]. Thus, it is a tenable hypothesis that this post prandial state underlies the vascular aging phenotype since endothelial culture studies suggest that physiological elevations in glucose and lipids induce oxidative stress and inflammation and also negatively alter “energy sensitive” pathways [4345]. Due to the lack of appropriate fasting time to resolve the acute metabolic insults, these alterations in apparently normal healthy middle aged and older adults may “set the table” for sustained endothelial dysfunction mediated by chronic vascular endothelial oxidative stress and inflammation. This viable hypothesis needs to be empirically tested, but would be consistent with the existing associative data.

3a. Aging, Nitric Oxide (NO) Bioavailability and Endothelial Function

In the vascular endothelium, L-arginine and the cofactors; BH4 and FADH, are necessary for the production of NO by the enzyme endothelial NO synthase (eNOS) [46]. NO is one of the most important vasodilatory and anti-atherosclerotic molecules produced by the endothelium [4648]. The major mechanism involved in the age-related reduction in EDD is reduced vascular NO bioavailability [49]. While initially thought to be the result of reduced expression of eNOS [5052], evidence from endothelial cells collected from human and animal arteries demonstrates that eNOS protein expression cannot explain the reduction in NO bioavailability with advancing age [19, 5355]. Rather, it appears that alterations in eNOS activation status, substrate/cofactor availability and/or inactivation of NO underlie the age-related reduction in NO bioavailability and impairments in EDD. Recent evidence also suggests that prostanoid vasodilators, such as prostacyclin, are diminished with advanced age [56, 57] and may explain some of the reductions in vasodilation. Furthermore, it is appreciated that reductions in NO promote enhanced vasoconstriction mediated by endothelial derived ET-1 [58, 59]. It has been shown that aging results in elevated expression of ET-1 and cyclooxgenase (producer of vasoconstrictor prostaglandins) [19, 58, 60, 61] and this elevation suppresses endothelial vasodilation in older adults and rodent models [19, 62]. Next we will delineate the two macro mechanistic processes which directly lead to suppression of endothelial function in older adults.

3b. Aging, Oxidative Stress and Endothelial Dysfunction

In humans, sedentary aging is considered a state of chronic, systemic “oxidative stress” [63] based primarily on observations of age-related elevations of plasma oxidative stress markers [21, 64, 65]. A key mechanism underlying age-associated reductions in EDD and NO bioavailability is the development of vascular oxidative stress [21, 49, 6669]. Oxidative stress can be defined as a state in which the bioavailability of reactive oxygen species (ROS) is increased relative to antioxidant defenses [7072]. Superoxide anion (O2) is a ROS produced by oxidant enzyme systems (NADPH oxidase-p67 phox, xanthine oxidase, cytochrome P450 2C9, uncoupled eNOS) [70, 71, 73, 74] that can inactivate NO [71, 75]. Interestingly, O2 can also be produced in abundance in the mitochondria at complex 1 and 3 [76]. However, mitochondrial O2 is released in the mitochondrial matrix (complex 1) or the intermembrane space (complex 3). Due to its highly reactive state, it is unlikely to navigate through multiple membranes to the cytosol in abundant quantities without being transformed to a less reactive form of ROS, such as hydrogen peroxide (H2O2). Nevertheless, O2 in the cytosol or extracellular space interacts with NO to produce peroxynitrite (ONOO), a ROS that itself nitrates cellular proteins forming nitrotyrosine, which can be used as a robust marker of cellular oxidative stress and ONOO production [7779]. Aging is associated with an increased abundance of arterial nitrotyrosine in animal models [29, 55, 80] and in arterial endothelial cells of humans [22]. In addition to the nitration of tyrosine which is irreversible, aged arteries also exhibit markers of lipid peroxidation such as 4-hydroxynonenal (4-HNE) or malodialdehyde (MDA) and/or other reversible regulatory oxidative stress marker glutathionylation at cysteine residues [8184]. Collectively, these oxidative stress-driven biochemical events result in a reduction in NO bioavailability and, subsequently, impaired EDD [66, 68].

Evidence of oxidative stress-associated suppression of NO and EDD in older arteries is supplied by the observations that acute administration of antioxidants, such as vitamin C and superoxide dismutase (SOD) mimetics, that reduce O2, nearly unilaterally improve NO bioavailability and EDD in older animals and humans [21, 66, 85]. An important source of increased vascular O2 and oxidative stress with aging appears to be NADPH oxidase [22, 55, 75]. Animal studies have shown similar improvement in EDD after blockade of NADPH oxidase as that seen with the use of SOD mimetics [55, 86]. Uncoupled eNOS is another important source of O2. eNOS uncoupling occurs when the critical cofactor BH4is inadequate, leading eNOS to produce O2 instead of NO [87]. Reducing uncoupled eNOS, by restoring BH4, improves EDD in older adults by reducing oxidative stress-mediated suppression of EDD [88]. This has been further supported by the observation of reduced O2 after BH4 administration in ex vivo arterioles from old rats [89]. In contrast, we have not found an aging-related increase in expression and/or activity of the other major oxidant enzymes (i.e., xanthine oxidase and cytochrome P450) in the aorta of mice or in vascular endothelial cells obtained from humans [22, 55, 90, 91]. Furthermore, studies using pharmacological inhibitors of xanthine oxidase and cytochrome P450 2C9 have resulted in no improvement in EDD in older adults [90, 92]. Interestingly, mitochondrial ROS has been shown to contribute to the ROS production/spillover in arteries from old rodents. However, this mitochondrial ROS is in the form of H2O2 and not directly as O2 [93] and is likely due to the production of O2 in the mitochondrial matrix. Still, targeting mitochondrial O2 reduces arterial O2, increases NO and improves EDD [94, 95]. This can be done either through genetic deletion of the mammalian Shc adaptor protein, p66shc (a protein that can increase mitochondrial H2O2 production and inhibit FoxO3a activity) or by introducing exogenous antioxidants targeted to the mitochondria, e.g. MitoQ.. Likewise, old MnSOD haploinsufficient mice have impaired EDD compared with old wildtype mice [96], supporting the importance of mitochondrial O2 in aged arteries. The mechanism of action of this phenomenon has yet to be elucidated, but does suggest that O2 produced in the mitochondria in excess indirectly alters NO, an effect that may result from the influence of H2O2 on cytosolic or extracellular O2 producers, such as NADPH oxidase [97] or from cycling of H2O2 into other ROS, such as Fenton reaction production of OH that then maybe able to inactivate NO.

The contribution of reduced antioxidant defenses against the development of systemic and vascular oxidative stress with sedentary aging is less clear. Circulating markers of antioxidants have been shown to be either reduced [64, 65] or unchanged with aging in humans [21]. Similarly, reduced expression of antioxidant enzymes, including intracellular and extracellular isoforms of SOD, have been observed in the vasculature of old animals [98, 99], although not in all cases [100, 101]. Furthermore, the concentration of glutathione peroxidase, a critical regulator of H2O2, has been shown to be similar between young and old arteries [96, 102] though mice which lack glutathione peroxidase have potentiated arterial dysfunction (both smooth muscle and endothelial) and oxidative stress in late adulthood (1 yr. old) [81]. Our human endothelial cell data suggests similar levels of SOD (CnZn & Mn) and catalase between populations of healthy older adults and young controls [22]. However, it has been reported that nitration of MnSOD may independently reduce its activity [103] and nitration of MnSOD is elevated in middle-aged and old rat aortas [53], unfortunately activity of Mn SOD was not measured. Still, in two independent studies we have demonstrated similar MnSOD activity from young and old mice in whole aortic homogenates [29, 104]. Therefore, it remains unclear if this nitration of Mn SOD has functional consequences in aged arteries or if Mn SOD activity is reduced in older endothelial cells. A summary age-related endothelial cell changes is provided in Figure 1. Taken together, these findings indicate that the expected increase in the expression of antioxidant enzymes that would be appropriate in response to increased ROS production with aging does not occur and thus an inappropriate transcriptional response to enhance antioxidant defenses likely contributes subsequent oxidative stress.

Figure 1. Age-associated Endothelial Oxidative Stress and Impaired NO Bioavailability.

Figure 1

In younger endothelial cells (Upper Panel), endothelial nitric oxide synthase (eNOS) has adequate cofactor availability, e.g., tetrahydrobiopterin (BH4), and produces nitric oxide (NO) through the conversion of L-arginine to L-citrulline. Reactive oxygen species (ROS), e.g., superoxide (O2) and hydrogen peroxide (H2O2), produced by the mitochondrial electron transport chain (ETC) or cytosolic oxidant enzymes, such as NADPH oxidase (NOX), are quenched by endogenous antioxidant enzymes (superoxide dismutase [SOD] and catalase). In older endothelial cells (Lower Panel), ROS produced in the mitochondria increase NOX mediated O2, this quenches NO bioavailability, through its conversion to peroxynitrite (ONOO), as well as uncouple eNOS by reducing BH4 availability. In the face of unchanged antioxidant defenses, these effects lead to a reduction in NO bioavailability and a pro-oxidant phenotype in the aged endothelium.

One caveat to the interpretation of the age-related finding above is that this excess O2 produced with aging is deleterious, whereas a normal amount of O2 production is essential for transcriptional signaling. Furthermore, O2 is actually essential for intact EDD in resistance arteries from young rodents and older rodents that have undergone exercise training or caloric restriction, as indicated by impaired EDD in response to SOD mimetics [86, 102, 105]. Likewise, antioxidants reduce brachial artery dilation to handgrip exercise in exercise-trained older adults [106]. Furthermore, some ROS, such as H2O2, are potent vasodilators that can compensate for reductions in NO [107, 108]. Thus, it is clear that ROS are necessary for proper arterial function, but when out of balance, can induce a critical dysfunction in the vascular endothelium.

3c. Aging, Inflammation and Endothelial Dysfunction

Initial observations made in circulating plasma suggested that aging was associated with chronic, low-grade inflammation characterized by increases in circulating acute phase proteins (e.g., C-reactive protein [CRP]) and pro-inflammatory cytokines [22, 91, 109], such as tumor necrosis factor alpha (TNF-α) [110] and interleukin (IL)-6 [22, 91, 111]. More recently, it has been demonstrated that expression of the pro-inflammatory cytokines TNF-α, IL-1β, IL-6 and interferon gamma (IFN-γ) are also elevated in the large elastic arteries of old mice [112, 113] and humans [114]. This age-associated pro-inflammatory arterial phenotype is downstream of increased nuclear factor κB (NFκB) activity. NFκB is a transcription factor that resides in the cytoplasm through its interaction with the inhibitory protein, I kappa B-alpha (IκB-α)[115, 116]. In response to inflammatory stimuli [116] or ROS [117119], I kappa B kinase beta (IKKβ) is activated and subsequently phosphorylates IκB-α, releasing its inhibition and allowing NFκB to translocate into the nucleus where it can activate gene transcription of pro-inflammatory cytokines [120]. Supporting the critical role of NFκB in age-related inflammation dependent endothelial dysfunction is evidence that pharmacological inhibition of NFκB signaling significantly reduces cytokines and enhances EDD in old mice and humans [113, 121, 122].

This pro-inflammatory arterial phenotype has been demonstrated in the vascular endothelial cells of older healthy humans [22, 91] and underlies endothelial dysfunction in older humans and mice [113, 121]. Evidence for a direct role of inflammation in endothelial dysfunction is provided by studies in which exogenous administration of pro-inflammatory cytokines was shown to induce endothelial dysfunction or endothelial activation in primary endothelial cells or isolated arteries [123125]. Such an inflammation-mediated endothelial dysfunction has also been observed in vivo in carotid arteries of aged rats [52] and in adipose tissue arteries of mice with diet-induced obesity [125], with the dysfunction occurring downstream of elevated TNF-α. In vivo arterial TNF-α may be produced locally by vascular cells [52, 112] or by immune cells infiltrating the adventitia of the large arteries [112]. TNF-α associated pro-inflammatory signaling may occur both up- and down-stream of elevated NFκB activity which itself underlies vascular dysfunction, at least in part, via increases in oxidative stress [52, 112, 113].

In addition to exacerbating inflammation downstream of NFκB transcription of pro-inflammatory cytokines, inflammatory signaling also stimulates O2 production and oxidative stress (and vice versa) through a number of mechanisms. These include increased NFκB mediated transcription of redox-sensitive genes like those encoding subunits of NADPH oxidase [126128] that increase ROS bioactivity and further activation of IKK-NFκB signaling. Thus, NFκB lies at the center of a vicious cycle that can exacerbate oxidative stress and inflammation (Figure 2). Interestingly, endothelial NFκB can impact the healthspan/lifespan beyond its effects on vascular function per se. Indeed, inhibition of endothelial NFκB signaling protects against not only age-associated vascular senescence and oxidative stress, but also protects against diet- and age-associated insulin resistance and increases lifespan in a mouse model expressing a dominant negative IKK in the endothelium [129].

Figure 2. Inflammation and Oxidative Stress in the Aged Endothelium: A Vicious Cycle.

Figure 2

The pro-inflammatory transcription factor, nuclear factor kappa B (NFκB) normally resides in the cytosol, where it is inactive, and endothelial nitric oxide synthase (eNOS) produces nitric oxide (NO) that is released from the endothelium and acts on the vascular smooth muscle to cause relaxation. With aging, the endothelial environment is perturbed by increases in cytokines and reactive oxygen species (ROS; e.g., superoxide [O2]), that can both be produced within the endothelium or by neighboring immune cells. These cytokines exacerbate oxidative stress and inflammation in the endothelium by activating oxidant enzymes, such as NADPH oxidase (NOX) increasing O2 production, as well as by acting in a feed forward manner to increase pro-inflammatory NFκB transcription. Likewise, the oxidative stress produced in the microenvironment of aged arteries, also acts in a feed forward manner to increase pro-inflammatory NFκB activity and activate neighboring immune cells as well as contributes directly to impaired NO by reducing eNOS activity via decreased BH4 availability as well as by quenching NO, leading to impaired endothelium dependent dilation in aged arteries. Thus, with aging there is a vicious cycle in aged arteries, in which inflammation and oxidative stress exacerbate one another impairing NO bioavailability and endothelial function.

In addition to NFκB signaling, there are a number of other inflammation-sensitive pathways that may also be involved in age-associated vascular dysfunction. Inflammatory and growth factor stimuli destabilize the vascular endothelium [130132] via activation of the small GTPase, ADP-ribosylation factor 6 (ARF6), its activator ARF nucleotide binding site opener (ARNO) and the downstream GTPase, Rac [132]. ARF6-ARNO-Rac act to reduce endothelial cell-cell interactions promoting vascular permeability, leukocyte adhesion and angiogenesis [132134]. Dysregulation of this inflammation-sensitive pathway can lead to disruption of normal endothelial barrier function, vascular leak, tissue disruption and pathological angiogenesis in retinal vascular disease [132, 134]. However, the impact of aging on this pathway and its role in vascular aging are unknown. Similarly, inflammatory signaling activates the Notch pathway to inhibit cell growth and promote hyper permeability and cell senescence (the implications of which will be discussed later in this review)[135, 136]. Interestingly, Notch signaling is enhanced in atherosclerotic regions of aortas from mice and humans and is activated in endothelial cells of older adults [135]. Notch activity is also increased in models of accelerated aging in response to progerin expression, a mutant “prelamin A” protein [137, 138]. Moreover, expression of the progerin protein is elevated in vascular tissues and cells from otherwise healthy aged humans [139, 140], suggesting that augmented Notch signaling may underlie at least some of the effects of vascular aging [141]. Thus, in addition to NFκB, other inflammation-sensitive pathways also have the potential to play a role in vascular dysfunction, e.g. impaired endothelial barrier function/vascular hyperpermeability [26, 31, 142, 143] and increases in cell senescence, all of which are associated with advancing age.

4. Cellular Senescence and Genomic Instability

4a. Cellular Senescence in Endothelial Aging

Cellular senescence, or permanent cell cycle arrest, is considered cellular aging, as it occurs in vitro after a certain number of cell cycles and in response to excessive intracellular and extracellular stressors [144, 145]. Senescence primarily occurs in the G0/G1 phase of the cell cycle and is a vital tumor suppressive mechanism that prevents passing damaged DNA to daughter cells or potential neoplastic transformation of damaged cells [144, 145]. Since being first described by Leonard Hayflick as an in vitro phenomenon in human fibroblasts, the potential role of senescence in in vivo aging and disease has been difficult to assess and somewhat controversial [146]. However, recent studies have shown that senescent cells accumulate in normal arterial tissue over the lifespan of humans [147, 148]. Likewise, the accumulation of senescent cells has been reported in diseased tissues, such as atherosclerotic plaques [149] and abdominal aortic aneurysms [150]. Baker et al. showed that clearance of senescent cells reversed aged and diseased phenotypes in a mouse model of accelerated aging [151]. This important study strongly suggested that there were phenotypic properties of senescent cells that were problematic to tissues, and potentially contribute to aging and chronic disease.

There are several causes of cellular senescence in mammalian cells, including excessive mitogenic signals [152], increases in extracellular or intracellular stressors like oxidative stress[153], chromatin disruptions [154], and DNA damage [155]. Senescence is induced by the two major tumor suppressor pathways, known as the p53 and p16/pRB pathways [156]. The p53 pathway depends on activation of the transcription factor p53 by a number of different signaling cascades [156]. One of the most important of these is the DNA damage response pathway [155, 157]. The p16/pRB pathway involves cyclin-dependent kinase inhibitors 2A (p16)-mediated inhibition of retinoblastoma-like protein 1 (pRB)[158]. The preference toward one pathway versus another appears to be cell type-specific [156, 159], with variation across species [160]. For example, telomere dysfunction can lead to activation of the p53 or p16/pRB pathway in human cells, but will only trigger the p53 pathway in rodent cells [160]. The general consensus seems to be that senescence via the p53 pathway is activated primarily by DNA damage and telomere dysfunction, while the p16/pRB pathway is linked primarily to mitogenic stress, chromatin disruptions, or general cellular stress [156, 159, 161].

In vitro senescent cells are characterized by a pro-inflammatory, pro-oxidative senescence-associated phenotype (SASP)[144]. The release of inflammatory mediators and ROS production likely reinforces cell cycle arrest in an autocrine fashion and activates immune cell surveillance of senescent cells [144, 162]. The SASP occurs within a few days of senescence induction in cells and appears to be irreversible due to stable chromatin modifications around clusters of SASP genes [163165]. The SASP profile in vascular endothelial cells from humans and rodents has been characterized by in vitro studies using comprehensive arrays of inflammatory cytokines and chemokines [144, 165169]. The release of IL-6, IL-1, IL-8, TNF-α, and monocyte chemoattractant protein-1 (MCP-1) has been linked to p53-mediated senescence in human vascular cells in vitro [170, 171]. Interestingly, P16/pRB-induced senescence has not been shown to lead to a SASP in human cells [144, 164]. Additionally, human arterial endothelial cells that had undergone replicative senescence in vitro exhibited elevated levels of H2O2 and O2 as well as reductions in NO [172, 173]. This SASP profile is possibly due to mitochondrial uncoupling or alterations in eNOS [172, 174]. Several lines of evidence support a major role for NFκB in the induction and maintenance of the p53-mediated SASP in human and rodent cells [175177]. Indeed, inhibition of NFκB allowed cells to escape p53-mediated senescence and reduced oxidative stress in rodents [175, 177]. This alludes to the importance of the SASP in reinforcing p53-mediated senescence and the importance of NFκB in promoting the SASP. Additionally, p53 activation leads to a shift from glycolytic to oxidative phosphorylation energy metabolism [178], which could result in increased flux through the electron transport chain and ROS release from mitochondria. Taken together endothelial cell senescence could be cause or consequence of the age-related increase in oxidative stress and likely contributes to the spread of vascular inflammation via the SASP, from senescent cells (Figure 3). Lastly this chronic inflammation could persist and accumulate, indefinitely in the vasculature until removal of the senescent cells, thus limiting therapeutic options.

Figure 3. Endothelial Senescence and Aging.

Figure 3

With advancing age, reactive oxygen species (ROS), as well as genotoxic stressors and telomere dysfunction, lead to double strand DNA breaks and genomic instability (Upper Panel). This genomic instability induces the DNA damage response, leading to the activation, p53 and nuclear factor kappa B (NFκB) that then transcribe genes that contribute to cell senescence such as the cyclin dependent kinase inhibitor, p21; oxidative stress, e.g., NADPH oxidase (NOX); and inflammatory cytokines. The cytokines and ROS act in a paracrine manner to impair function in neighboring endothelial cells (Lower Panel).

4b. Aging, Genomic Instability, Cellular Senescence and Endothelial Dysfunction

Genomic instability, or DNA damage acquired over time, is an important mechanism that may underlie the age-related accumulation of senescent cells reported in arterial tissues [147, 148]. Age-related genomic instability in vascular cells can occur from a variety of genotoxic insults, including oxidative stress [179, 180] and mechanical stress [181]. DNA damage results in temporary cell cycle arrest to allow DNA repair pathways time to repair breaks prior to a cell entering S phase during replication [155]. If damage is persistent or extensive, permanent cell cycle arrest or even apoptosis will ensue [155]. Common forms of DNA damage relevant to aging include single and double strand DNA breaks and DNA adducts. While the age-related accumulation of DNA breaks and DNA adducts have not been shown in human vascular cells, DNA breaks have been shown to occur with advancing age in sperm cells [182] and the mitochondrial DNA of skeletal muscle [183, 184]. Microsatellite instability and loss of heterozygosity, which is thought to be a consequence of DNA breaks, has been linked to pulmonary artery hypertension [185] and atherosclerosis [186191] in humans. Interestingly, two recently developed mouse models of disrupted DNA adduct repair (xeroderma pigmentosum D (XPD)ttd and ERCC1d/− mice) demonstrated age-related impairments in EDD in response to acetylcholine, reduced eNOS expression and activity, and increased p53 expression and ROS content [192].

Telomere dysfunction is another form of genomic instability that may lead to cellular senescence with advancing age. First described by Harley et al., senescence triggered by replication-dependent telomere dysfunction is often referred to as replicative senescence [193]. Telomeres can be damaged by the genotoxic stressors described above [194, 195], and in vitro studies in various human cell types have clearly shown that the breakdown of telomere structure, referred to as telomere uncapping, leads to p53 activation and senescence [196, 197]. The only previous study to measure arterial telomere uncapping reported greater uncapping with advancing age, which was linked to p53-mediated senescence [148].

A vicious cycle of oxidative stress leading to DNA damage or telomere uncapping-mediated p53 activation and subsequent senescence-associated oxidative stress is a simple model of the role of genomic instability in age-related endothelial dysfunction. However, a more integrative model might consider the influence of p53-mediated shifts in energy metabolism on the following [178] energy sensitive pathways: SIRT-1, AMPK and mTOR that are known to influence both aging and endothelial function and that are discussed next.

5. Energy Sensing Longevity Pathways

5a. SIRT-1, Aging and Endothelial Dysfunction

Sirtuins were originally discovered in a screen for gene silencing factors in yeast and, therefore, given the name Sir2 (silent information regulator 2). Little research was conducted on the Sir2 family until Guarente et al. identified these proteins as critical regulators of longevity [198200]. Thereafter, Sir2 and its mammalian homologues, the sirtuin family (SIRT1-7), of NAD+-dependent protein deacetylases and ADP-ribosyltransferases were quickly identified. In mammals, SIRT1-4 have been implicated in the control of cellular metabolism with SIRT-2, 3 and 4 predominantly expressed in the mitochondria and SIRT-1 expressed in the nucleus, with some cytoplasmic expression [201203]. The sirtuin family and specifically SIRT-1 are implicated in a majority of the physiological benefits of caloric restriction [203205]. SIRT-1 expression decreases in a multitude of tissues with advancing age [206208]. SIRT-1 function is related to deacetylation and thereby modulating activity of nuclear transcription factors, co-regulators and proteins to adapt gene expression in response to the cellular energy state and provide “stress resistance” by reducing pro-inflammatory and oxidative stress pathways [209]. Interestingly, activation of SIRT-1 with the small molecule SRT1720 increases lifespan and preserves glucose tolerance in rodents [210] Taken together these data suggest that SIRT-1 activation may have significant promise for improving endothelial function with aging or CVD.

SIRT-1 has been reported to modulate NO and endothelial function in small and large arteries directly via deacetylation and subsequent activation of eNOS [207, 211]. Our group has demonstrated that SIRT-1 expression and activity decrease with age in the vasculature in both mice and humans [104, 207, 212, 213], and this is associated with age-related endothelial dysfunction in mice [104, 207]. Interestingly in rodent models, both short and long term caloric restriction can prevent the decline in arterial SIRT-1 and endothelial function [104, 212, 214]. Most of the data related to the vascular endothelium and SIRT-1 activators is restricted to studies utilizing resveratrol, a naturally occurring polyphenol [215]. Unfortunately, resveratrol must be used at high doses to activate SIRT-1 [216] and is known to activate over 15 unique pathways in addition to being a potent antioxidant and phytoestrogen [215, 217221]. While these characteristics of resveratrol do not alter its potential therapeutic value, they do generate some difficulty in elucidating its mechanism of action, specifically whether SIRT-1 activation is responsible for its beneficial effects. It has been shown that very high doses of resveratrol (2400 mg/kg of food) improves EDD in isolated arteries from middle-aged animals after 1 year of treatment [222], although it is unknown by what mechanism(s) this effect was achieved. Subsequently, we have utilized SRT-1720 treatment in old mice and have demonstrated that four weeks of SRT-1720 normalizes SIRT-1 activity and reduces age-related NFκB acetylation, arterial inflammation and oxidative stress [213, 223]. Somewhat surprisingly SRT-1720 improvements in EDD were not mediated by increased NO bioavailabilty in old mice, but rather via augmented COX-2 vasodilators [213, 223]. Therefore, despite strong evidence suggesting a role of reduced SIRT-1 in the age-related reduction in NO bioavailability and endothelial dysfunction, treatment with a SIRT-1 activator improves endothelial function and reduced oxidative stress and inflammation, but does not restore NO. Future studies will be needed using small molecule activators of sirtuins to determine if they can ameliorate age-related CVD pathologies.

5b. AMP-activated protein kinase (AMPK), Aging and Endothelial Dysfunction

AMPK, a highly conserved heterotrimeric serine-threonine kinase, is an important energy sensing signaling protein that integrates energy balance, metabolism and stress resistance [224, 225]. AMPK is activated in response to increases in the AMP:ATP ratio and following physiological stimuli such as shear stress, heat shock, exercise and hypoxia [226]. AMPK is made up of a catalytic α subunit, a structural β subunit and the AMPK binding site containing γ subunit, each of which exist in multiple isoforms [226]. Activation of AMPK requires phosphorylation of the α subunit and occurs downstream of two kinases; LKB1 and Ca2+/calmodulin-dependent protein kinase kinase β (CaMKKβ) (reviewed elsewhere [226, 227]). Shear stress-induced AMPK activation in endothelial cells is independent of Akt and involves the activation of the α2 subunit via LKB1 [226]. AMPK activation can also be mediated pharmacologically with aminoimidazole carboxamide ribonucleotide (AICAR) or metformin. AICAR is a direct activator of AMPK that acts downstream of LKB1 leading to phosphorylation of the α2 subunit of AMPK [226]. In contrast, metformin is an indirect activator of AMPK [228], actions of which are mediated by an increase in the AMP:ATP ratio resulting from inhibition of Complex I of the respiratory chain [229, 230]. However, such inhibition of oxidative phosphorylation, and a subsequent increase in glycolysis, can have numerous effects independent of AMPK, e.g., metformin effects transcription factors and kinases involved in numerous cell cycle and metabolic pathways (p53, p38, MAPK, PKC and Akt)[231234]. Therefore, as with resveratrol, metformin and other non-specific agents are not optimal for mechanistic studies evaluating these pathways but do afford insights as potential therapeutic agents.

AMPK activation in vascular endothelial cells can contribute to both angiogenesis and NO production. Angiogenic effects of AMPK occur both upstream and downstream of vascular endothelial growth factor (VEGF), with the effects of AMPK signaling promoting differentiation of endothelial progenitor cells [235] as well as angiogenesis in isolated myocardial microvascular endothelial cells [236], in ischemic skeletal muscle [237] and during hypoxia both in vitro and in vivo [238]. AMPK activation also results in increased activation of eNOS [239, 240] via signaling through Rac1 and Akt [238, 240], as well as through direct phosphorylation of eNOS [241, 242]. While the pro-angiogenic effects of AMPK have been linked to increased VEGF expression [236, 237], AMPK has also been demonstrated to transcriptionally regulate a number of other proteins involved in inflammation, mitochondrial biogenesis, fatty acid and cholesterol synthesis, glucose metabolism, cell growth and oxidative stress signaling [226], all of which may impact vascular function. Taken together, these findings suggest that augmenting AMPK signaling may not only enhance angiogenesis and increase bioavailability of NO and vasodilatory responses [235, 238, 240], but may also impact upstream mediators of age-associated endothelial dysfunction, such as inflammation and oxidative stress. Indeed, AMPK activation improves endothelial function in type I and II diabetic rodents [243245] but, less is known about the activation state of AMPK in aged arteries or its role in age-associated vascular dysfunction.

In the context of vascular aging, although there is a report of increased AMPK activation in endothelial cells cultured to senescence, a cellular model of aging [246], AMPK activity is reduced in the aorta[247] and cerebral arteries [248] of old rodents. Furthermore, the pharmacological activation of AMPK by AICAR increases EDD in old mice [247], suggesting that inactivation of arterial AMPK contributes to age-associated endothelial dysfunction. However, despite evidence for AMPK-mediated eNOS activation [238, 240242], the effect of in vivo AMPK activation by AICAR to improve dilation in arteries from old mice was not mediated by an increased NO bioavailability [247]. Similar NO independent effects of AICAR were also found after acute in vitro administration to isolated aortic rings, in which AICAR induced relaxation of agonist constricted vessels independent of NO [249]. Similar to treatment of old mice with SIRT-1 activators, AMPK activators therefore reduce arterial oxidative stress and improve endothelial function, but do so in an NO independent mechanism.

5c. Mammalian target of rapamycin (mTOR), Aging and Endothelial Dysfunction

Rapamycin was discovered more than 30 years ago from an Easter Island soil sample. It is a potent antifungal metabolite produced from bacteria and in addition to its antifungal properties was quickly found to have antiproliferative and immunosuppressant properties when used in high quantities. The mammalian target of rapamycin (mTOR) is a signaling protein which responds to nutrients (i.e. amino acids) or growth factors (i.e. insulin) in order to modulate mRNA translation, protein synthesis and cellular growth [250]. mTOR signaling is composed of two distinct pathways the mTOR complex 1 (mTORC1), the more typically studied and rapamycin “sensitive”, and mTOR complex 2 (mTORC2), the less clearly identified and less sensitive to rapamycin. Most information to date on the role of mTOR has studied the insulin/nutrient signaling via the mTORC1 and significantly less in known about the role of mTORC2 (in this review, future references measure either mTORC1 or general mTOR activity)[251]. Earlier this decade studies showed that decreasing TOR signaling, genetically or with rapamycin, in yeast, Drosophila, and C. elegans is able to slow aging and increase lifespan [252255]. Follow-up studies out of Richard Miller’s laboratory reproduced these findings in mice fed a diet with rapamycin incorporated [256, 257]. These studies suggested that inhibiting mTOR via rapamycin could delay age-associated diseases and extend lifespan in mammals. A subsequent study replicated these findings by genetically manipulating a downstream target of mTOR, ribosomal S6 protein kinase (S6K1) and demonstrated a similar phenotype of reduced risk factors for age-related diseases (bone density, insulin sensitivity) and increase lifespan [258]. These studies unilaterally implicate elevated mTOR signaling in accelerated aging and associated diseases. Currently, very little is known about mTOR and endothelial function. We have demonstrated that mTOR signaling is augmented in arteries from older mice which display endothelial dysfunction and lifelong caloric restriction prevents augmented arterial mTOR signaling and endothelial dysfunction [104]. Furthermore, preliminary studies in our lab suggest that 6–8 weeks of dietary supplementation of rapamycin improve NO and endothelial function in old mice [259]. Chronic rapamycin and other rapamycin analog studies are warranted to determine if inhibition of the mTOR pathway is a viable method to improve endothelial function in older adults.

6. Interactions of mTOR, SIRT1 and AMPK: Role in the Vascular Aging Phenotype

Although the energy sensor pathways, SIRT-1, AMPK activation and mTOR inhibition have been linked to the vascular aging phenotype, when viewed from a wider perspective, what becomes evident is that these pathways do not, in fact, act in parallel to independently impact the macromechanistic processes (e.g. inflammation, oxidative stress and senescence) associated with vascular dysfunction. Rather, there are points of convergence and crosstalk among these pathways that not only impact the downstream effects, but also the expression of the signaling molecules themselves. Indeed, signaling through the transcription factors NFκB, FoxO and p53, represent points of convergence for signaling downstream of mTOR, SIRT-1 and AMPK.

6a. NFκB

The acetylation of NFκB p65 subunit by p300 prevents the nuclear export of NFκB, while its deacetylation by SIRT-1 allows for the association of NFκB with IκB-α and its subsequent nuclear export and inactivation [260]. Inhibition of mTOR (complex 1) by rapamycin inhibits phosphorylation of NFκB and subsequent nuclear translocation via its influence on IKKβ activity [261263]. AMPK signaling has a complex interaction with NFκB [264], such that AMPK can both indirectly inhibit pro-inflammatory NFκB signaling [265267] and activate NFκB signaling leading to its anti-apoptotic effects in endothelial cells [268]. Taken together these studies indicate that SIRT-1, mTOR and AMPK can all alter the pro-inflammatory transcription factor NFκB activity by both distinct and intersecting pathways. It also indicates that reductions in AMPK and SIRT-1 activity and conversely increased mTOR activity may increase NFκB signaling resulting in a pro-inflammatory phenotype similar to that observed in older arteries

6b. FoxO transcription factors

Fox nuclear transcription factors are a family of proteins characterized by a conserved 100 amino-acid sequence known as the “forkhead box.” The FoxO subgroup have been implicated in ROS detoxification, cell cycle regulation, apoptosis, and DNA repair [269]. The activity of this subgroup is dependent on posttranslational modifications, such as phosphorylation and acetylation. FoxO proteins are constitutively held in the cytoplasm, and phosphorylation by signaling kinases inhibit their transcriptional activity by preventing nuclear translocation [270]. Phosphorylated FoxO is retained in the cytoplasm by the 14-3-3 chaperone protein, and phosphorylation at serine residues 256/253 [271] targets FoxO proteins for degradation via ubiquitination [272]. The downstream targets of nuclear FoxO activity are determined by acetylation [273]. For example, p300-mediated FoxO3a acetylation induces the preferential transcription of pro-apoptotic gene targets. SIRT-1 deacetylates FoxO3a, leading to the preferential transcription of genes related to stress resistance, such as the antioxidants MnSOD and catalase [273275]. Furthermore, the FoxO4 isoform is an endogenous NFκB inhibitor, whose activity is enhanced when bound to SIRT-1 [276]. The interaction of mTOR and FoxO signaling occurs through modulation of other signaling kinases, such as protein kinase B (Akt) and serum- and glucocorticoid-regulated kinase-1 (SGK1). The phosphorylation of FoxO proteins by these kinases leads to the cytoplasmic retention and inactivation of FoxO transcription factors [277]. Rapamycin, an inhibitior of mTOR, can inhibit SGK1 activity downstream of the mTORC1 complex [278] and may, as a result, increase FoxO-mediated transcription of anti-oxidant gene targets. AMPK can also directly phosphorylate the 3a isoform of FoxO leading to its activation and subsequent inhibition of NFκB [279].

6c. p53

p53 is a transcription factor that acts as a tumor suppressor preventing the survival of malignant cells. p53 is activated by stress such as DNA damage, telomere attrition and hypoxia and leads to cell cycle arrest and apoptosis [280]. Endothelial dysfunction in older adults is associated with telomere dysfunction and subsequent increases in p53-mediated cell cycle arrest via increases in the cyclin dependent kinase inhibitor, p21 expression [148]. Furthermore, recent evidence points to a link between the DNA damage response and energy sensitive pathways via SIRT-1 activity and expression. SIRT-1 is involved in the DNA repair and its activity has been shown to protect human and rodent vascular cells from DNA damage in the context of atherosclerosis [281]. Thus, age-related reductions in endothelial SIRT-1 expression and activity [207] may be partly the result of depleted protein levels caused by increased need for repair of accumulating DNA damage. Conversely, reductions in endothelial SIRT-1 expression may lead to genomic instability with advancing age. Specifically SIRT-1 deacetylates p53 resulting in its inactivation and the promotion of cell survival, thus reductions in SIRT-1 activity with aging may contribute to p53 activation and increased cell senescence [282, 283]. Conversely, AMPK phosphorylates p53 on ser15 and ser20 [284, 285] leading to p53 stabilization, activation and cell cycle arrest [286]. Phosphorylation of p53 at these sites has also been associated with reductions in inflammation [284]. Although, AMPK induced decreases in cell senescence does not likely contribute to the aging vascular phenotype, a loss of the p53-associated anti-inflammatory effects of AMPK may be an important factor. The crosstalk between p53 and mTOR occurs in the inverse manner as described for SIRT-1 and AMPK, with p53 inhibiting mTOR activity [287]. However, as both p53 and mTOR activation is increased with vascular aging, it is unlikely that this inhibitory interaction is a significant contributor to the vascular aging phenotype.

7. Conclusions

Since the initial observations of endothelial dysfunction in older adults in the early 1990s, the field has achieved several significant milestones. First, due to initial work and reviews by early researchers, vascular aging now is appreciated as a critical step in the development of age-related CVD. This recognition has allowed for funding and expansion of the field of study. Second, the vascular aging research community has expanded dramatically in the last 20 years and has established viable primate and rodent models that phenocopy human aging. These models have allowed for greater mechanistic insight into the cellular and molecular mechanisms of endothelial dysfunction as well as the translation of interventions to human endothelial physiology studies. Third, in the past 25 years, the research community has identified multiple pathways that directly influence oxidative stress and inflammation, the major mechanisms of endothelial dysfunction with advancing age. Emerging evidence has suggested several promising pathways that could be viable candidates to reverse age-related endothelial dysfunction that require further exploration in areas outside of endothelial dilation; such as fibrinolysis, permeability and angiogenesis. Unfortunately, it is still unknown what the initiating events are that alter the cellular machinery to induce the well described vicious cycle of oxidative stress and inflammation. Indeed, although deregulation of the energy sensing pathways, e.g., SIRT-1, mTOR and AMPK, are attractive possible mechanisms, to date, pharmacological interventions that restore normal pathway activity in older animals have proven only partially effective at restoring endothelial function to that of young animals. Finally, treating older adults with prescription drugs to improve endothelial function as a preventative measure against CVD is not currently acceptable in the absence of clinical disease. Unless the definition of “clinical disease” is extended to include endothelial dysfunction, the only viable preventative interventions are those involving lifestyle changes or treatments with nutraceuticals. Overall, despite impressive advancements in our understanding of the biology of aging in the endothelium, we as a research community still have fundamental questions to answer including: What are the initiating events that lead to the arterial aging phenotype? And if a target pathway can be identified and viable drugs created to improve endothelial dysfunction in older adults, when should such therapy be initiated?

Research Highlights.

  • Aging and endothelial dysfunction are risk factors for cardiovascular disease

  • Both oxidative stress and inflammation suppress endothelial function in older adults

  • Genomic instability and senescence are present in older arteries

  • Dysregulated energy sensing pathways contribute to the age-related endothelial phenotype

  • The initiating events leading to age-related endothelial dysfunction are still unknown

Acknowledgments

We would like to acknowledge the help of Grant Henson, Phil Gates, Dan Machin, Dan Trott and Sarah Breevort for selected literature reviews and critical review of the manuscript. We would like to acknowledge Matt Holmes for his assistance in design and editing of the figures. This manuscript was supported by NIH grants: NIA R01 AG040297 (Donato), R21 AG043952 (Lesniewksi/Donato), K02 AG045339 (Donato) K01 AG029337(Walker), NIEHS T32 ES007032 (Morgan) and VA merit 1I01BX002151 (Lesniewksi)

Footnotes

Disclosures. The authors have no disclosures or conflicts of interest.

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

Literature Cited

  • 1.Health, United States, 2010: With Special Feature on Death and Dying. Hyattsville (MD): 2011. [PubMed] [Google Scholar]
  • 2.Lakatta E, Levy D. Arterial and cardiac aging: major shareholders in cardiovascular disease enterprises: Part I: aging arteries: a “set up” for vascular disease. Circulation. 2003;107:139–46. doi: 10.1161/01.cir.0000048892.83521.58. [DOI] [PubMed] [Google Scholar]
  • 3.Widlansky ME, Gokce N, Keaney JF, Jr, Vita JA. The clinical implications of endothelial dysfunction. J Am Coll Cardiol. 2003;42:1149–60. doi: 10.1016/s0735-1097(03)00994-x. [DOI] [PubMed] [Google Scholar]
  • 4.Franklin SS, Khan SA, Wong ND, Larson MG, Levy D. Is pulse pressure useful in predicting risk for coronary heart Disease? The Framingham heart study. Circulation. 1999;100:354–60. doi: 10.1161/01.cir.100.4.354. [DOI] [PubMed] [Google Scholar]
  • 5.Franklin SS, Larson MG, Khan SA, Wong ND, Leip EP, Kannel WB, et al. Does the relation of blood pressure to coronary heart disease risk change with aging? The Framingham Heart Study. Circulation. 2001;103:1245–9. doi: 10.1161/01.cir.103.9.1245. [DOI] [PubMed] [Google Scholar]
  • 6.Seals DR, Jablonski KL, Donato AJ. Aging and vascular endothelial function in humans. Clin Sci (Lond) 2011;120:357–75. doi: 10.1042/CS20100476. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Lakatta EG, Wang M, Najjar SS. Arterial aging and subclinical arterial disease are fundamentally intertwined at macroscopic and molecular levels. Med Clin North Am. 2009;93:583–604. doi: 10.1016/j.mcna.2009.02.008. Table of Contents. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Nagai Y, Metter EJ, Earley CJ, Kemper MK, Becker LC, Lakatta EG, et al. Increased carotid artery intimal-medial thickness in asymptomatic older subjects with exercise-induced myocardial ischemia. Circulation. 1998;98:1504–9. doi: 10.1161/01.cir.98.15.1504. [DOI] [PubMed] [Google Scholar]
  • 9.Celermajer DS, Sorensen KE, Spiegelhalter DJ, Georgakopoulos D, Robinson J, Deanfield JE. Aging is associated with endothelial dysfunction in healthy men years before the age-related decline in women. Journal of the American College of Cardiology. 1994;24:471–6. doi: 10.1016/0735-1097(94)90305-0. [DOI] [PubMed] [Google Scholar]
  • 10.Vaitkevicius PV, Fleg JL, Engel JH, O’Connor FC, Wright JG, Lakatta LE, et al. Effects of age and aerobic capacity on arterial stiffness in healthy adults. Circulation. 1993;88:1456–62. doi: 10.1161/01.cir.88.4.1456. [DOI] [PubMed] [Google Scholar]
  • 11.Asai K, Kudej RK, Shen YT, Yang GP, Takagi G, Kudej AB, et al. Peripheral vascular endothelial dysfunction and apoptosis in old monkeys. Arteriosclerosis, thrombosis, and vascular biology. 2000;20:1493–9. doi: 10.1161/01.atv.20.6.1493. [DOI] [PubMed] [Google Scholar]
  • 12.Li Z, Froehlich J, Galis ZS, Lakatta EG. Increased expression of matrix metalloproteinase-2 in the thickened intima of aged rats. Hypertension. 1999;33:116–23. doi: 10.1161/01.hyp.33.1.116. [DOI] [PubMed] [Google Scholar]
  • 13.Haudenschild CC, Prescott MF, Chobanian AV. Aortic endothelial and subendothelial cells in experimental hypertension and aging. Hypertension. 1981;3:I148–53. doi: 10.1161/01.hyp.3.3_pt_2.i148. [DOI] [PubMed] [Google Scholar]
  • 14.Franklin SS, Gustin Wt, Wong ND, Larson MG, Weber MA, Kannel WB, et al. Hemodynamic patterns of age-related changes in blood pressure. The Framingham Heart Study. Circulation. 1997;96:308–15. doi: 10.1161/01.cir.96.1.308. [DOI] [PubMed] [Google Scholar]
  • 15.RB, IF Vascular endothelium and blood flow. Handb Exp Pharmacol. 2006;176:43. doi: 10.1007/3-540-36028-x_2. [DOI] [PubMed] [Google Scholar]
  • 16.Pober JS, Min W, Bradley JR. Mechanisms of Endothelial Dysfunction, Injury, and Death. Annual Review of Pathology: Mechanisms of Disease. 2009;4:71–95. doi: 10.1146/annurev.pathol.4.110807.092155. [DOI] [PubMed] [Google Scholar]
  • 17.Lakatta EG, Levy D. Arterial and Cardiac Aging: Major Shareholders in Cardiovascular Disease Enterprises: Part I: Aging Arteries: A “Set Up” for Vascular Disease. Circulation. 2003;107:139–46. doi: 10.1161/01.cir.0000048892.83521.58. [DOI] [PubMed] [Google Scholar]
  • 18.Mitchell GF, Parise H, Vita JA, Larson MG, Warner E, Keaney JF, Jr, et al. Local Shear Stress and Brachial Artery Flow-Mediated Dilation: The Framingham Heart Study. Hypertension. 2004;44:134–9. doi: 10.1161/01.HYP.0000137305.77635.68. [DOI] [PubMed] [Google Scholar]
  • 19.Donato AJ, Gano LB, Eskurza I, Silver AE, Gates PE, Jablonski K, et al. Vascular endothelial dysfunction with aging: endothelin-1 and endothelial nitric oxide synthase. Am J Physiol Heart Circ Physiol. 2009;297:H425–32. doi: 10.1152/ajpheart.00689.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.DeSouza C, Shapiro L, Clevenger C, Dinenno F, Monahan K, Tanaka H, et al. Regular aerobic exercise prevents and restores age-related declines in endothelium-dependent vasodilation in healthy men. Circulation. 2000;102:1351–7. doi: 10.1161/01.cir.102.12.1351. [DOI] [PubMed] [Google Scholar]
  • 21.Eskurza I, Monahan KD, Robinson JA, Seals DR. Effect of acute and chronic ascorbic acid on flow-mediated dilatation with sedentary and physically active human ageing. J Physiol. 2004;556:315–24. doi: 10.1113/jphysiol.2003.057042. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Donato AJ, Eskurza I, Silver AE, Levy AS, Pierce GL, Gates PE, et al. Direct evidence of endothelial oxidative stress with aging in humans: relation to impaired endothelium-dependent dilation and upregulation of nuclear factor-kappaB. Circ Res. 2007;100:1659–66. doi: 10.1161/01.RES.0000269183.13937.e8. [DOI] [PubMed] [Google Scholar]
  • 23.Gerhard M, Roddy M, Creager S, Creager M. Aging progressively impairs endothelium-dependent vasodilation in forearm resistance vessels of humans. Hypertension. 1996;27:849–53. doi: 10.1161/01.hyp.27.4.849. [DOI] [PubMed] [Google Scholar]
  • 24.Taddei S, Galetta F, Virdis A, Ghiadoni L, Salvetti G, Franzoni F, et al. Physical activity prevents age-related impairment in nitric oxide availability in elderly athletes. Circulation. 2000;101:2896–901. doi: 10.1161/01.cir.101.25.2896. [DOI] [PubMed] [Google Scholar]
  • 25.Smith DT, Hoetzer GL, Greiner JJ, Stauffer BL, DeSouza CA. Effects of ageing and regular aerobic exercise on endothelial fibrinolytic capacity in humans. J Physiol. 2003;546:289–98. doi: 10.1113/jphysiol.2002.027870. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Blau CW, Cowley TR, O’Sullivan J, Grehan B, Browne TC, Kelly L, et al. The age-related deficit in LTP is associated with changes in perfusion and blood-brain barrier permeability. Neurobiol Aging. 2012;33:1005.e23–35. doi: 10.1016/j.neurobiolaging.2011.09.035. [DOI] [PubMed] [Google Scholar]
  • 27.Croley AN, Zwetsloot KA, Westerkamp LM, Ryan NA, Pendergast AM, Hickner RC, et al. Lower capillarization, VEGF protein, and VEGF mRNA response to acute exercise in the vastus lateralis muscle of aged vs. young women. J Appl Physiol (1985) 2005;99:1872–9. doi: 10.1152/japplphysiol.00498.2005. [DOI] [PubMed] [Google Scholar]
  • 28.Ryan NA, Zwetsloot KA, Westerkamp LM, Hickner RC, Pofahl WE, Gavin TP. Lower skeletal muscle capillarization and VEGF expression in aged vs. young men. J Appl Physiol (1985) 2006;100:178–85. doi: 10.1152/japplphysiol.00827.2005. [DOI] [PubMed] [Google Scholar]
  • 29.Lesniewski LA, Connell ML, Durrant JR, Folian BJ, Anderson MC, Donato AJ, et al. B6D2F1 Mice are a suitable model of oxidative stress-mediated impaired endothelium-dependent dilation with aging. J Gerontol A Biol Sci Med Sci. 2009;64:9–20. doi: 10.1093/gerona/gln049. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Yamamoto K, Takeshita K, Shimokawa T, Yi H, Isobe K, Loskutoff DJ, et al. Plasminogen activator inhibitor-1 is a major stress-regulated gene: implications for stress-induced thrombosis in aged individuals. Proc Natl Acad Sci U S A. 2002;99:890–5. doi: 10.1073/pnas.022608799. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Pelegri C, Canudas AM, del Valle J, Casadesus G, Smith MA, Camins A, et al. Increased permeability of blood-brain barrier on the hippocampus of a murine model of senescence. Mech Ageing Dev. 2007;128:522–8. doi: 10.1016/j.mad.2007.07.002. [DOI] [PubMed] [Google Scholar]
  • 32.Zhuo Y, Li SH, Chen MS, Wu J, Kinkaid HY, Fazel S, et al. Aging impairs the angiogenic response to ischemic injury and the activity of implanted cells: combined consequences for cell therapy in older recipients. J Thorac Cardiovasc Surg. 2010;139:1286–94. 94 e1–2. doi: 10.1016/j.jtcvs.2009.08.052. [DOI] [PubMed] [Google Scholar]
  • 33.Taddei S, Virdis A, Mattei P, Ghiadoni L, Gennari A, Fasolo CB, et al. Aging and endothelial function in normotensive subjects and patients with essential hypertension. Circulation. 1995;91:1981–7. doi: 10.1161/01.cir.91.7.1981. [DOI] [PubMed] [Google Scholar]
  • 34.Walker AE, Eskurza I, Pierce GL, Gates PE, Seals DR. Modulation of vascular endothelial function by low-density lipoprotein cholesterol with aging: influence of habitual exercise. Am J Hypertens. 2009;22:250–6. doi: 10.1038/ajh.2008.353. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.DeVan AE, Eskurza I, Pierce GL, Walker AE, Jablonski KL, Kaplon RE, et al. Regular aerobic exercise protects against impaired fasting plasma glucose-associated vascular endothelial dysfunction with aging. Clin Sci (Lond) 2013;124:325–31. doi: 10.1042/CS20120291. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Jablonski KL, Gates PE, Pierce GL, Seals DR. Low dietary sodium intake is associated with enhanced vascular endothelial function in middle-aged and older adults with elevated systolic blood pressure. Ther Adv Cardiovasc Dis. 2009;3:347–56. doi: 10.1177/1753944709345790. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Walker AE, Seibert SM, Donato AJ, Pierce GL, Seals DR. Vascular endothelial function is related to white blood cell count and myeloperoxidase among healthy middle-aged and older adults. Hypertension. 2010;55:363–9. doi: 10.1161/HYPERTENSIONAHA.109.145870. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Kaplon RE, Walker AE, Seals DR. Plasma norepinephrine is an independent predictor of vascular endothelial function with aging in healthy women. J Appl Physiol (1985) 2011;111:1416–21. doi: 10.1152/japplphysiol.00721.2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Basu R, Dalla Man C, Campioni M, Basu A, Klee G, Toffolo G, et al. Effects of age and sex on postprandial glucose metabolism: differences in glucose turnover, insulin secretion, insulin action, and hepatic insulin extraction. Diabetes. 2006;55:2001–14. doi: 10.2337/db05-1692. [DOI] [PubMed] [Google Scholar]
  • 40.Issa JS, Diament J, Forti N. Postprandial lipemia: influence of aging. Arq Bras Cardiol. 2005;85:15–9. doi: 10.1590/s0066-782x2005001400004. [DOI] [PubMed] [Google Scholar]
  • 41.Marchesi S, Lupattelli G, Schillaci G, Pirro M, Siepi D, Roscini AR, et al. Impaired flow-mediated vasoactivity during post-prandial phase in young healthy men. Atherosclerosis. 2000;153:397–402. doi: 10.1016/s0021-9150(00)00415-9. [DOI] [PubMed] [Google Scholar]
  • 42.Rudolph TK, Ruempler K, Schwedhelm E, Tan-Andresen J, Riederer U, Boger RH, et al. Acute effects of various fast-food meals on vascular function and cardiovascular disease risk markers: the Hamburg Burger Trial. Am J Clin Nutr. 2007;86:334–40. doi: 10.1093/ajcn/86.2.334. [DOI] [PubMed] [Google Scholar]
  • 43.Wu Y, Song P, Xu J, Zhang M, Zou MH. Activation of protein phosphatase 2A by palmitate inhibits AMP-activated protein kinase. J Biol Chem. 2007;282:9777–88. doi: 10.1074/jbc.M608310200. [DOI] [PubMed] [Google Scholar]
  • 44.Chakrabarti S, Davidge ST. High glucose-induced oxidative stress alters estrogen effects on ERalpha and ERbeta in human endothelial cells: reversal by AMPK activator. J Steroid Biochem Mol Biol. 2009;117:99–106. doi: 10.1016/j.jsbmb.2009.07.007. [DOI] [PubMed] [Google Scholar]
  • 45.Mortuza R, Chen S, Feng B, Sen S, Chakrabarti S. High glucose induced alteration of SIRTs in endothelial cells causes rapid aging in a p300 and FOXO regulated pathway. PLoS One. 2013;8:e54514. doi: 10.1371/journal.pone.0054514. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Alp NJ, Channon KM. Regulation of endothelial nitric oxide synthase by tetrahydrobiopterin in vascular disease. Arterioscler Thromb Vasc Biol. 2004;24:413–20. doi: 10.1161/01.ATV.0000110785.96039.f6. [DOI] [PubMed] [Google Scholar]
  • 47.Ganz P, Vita JA. Testing endothelial vasomotor function: nitric oxide, a multipotent molecule. Circulation. 2003;108:2049–53. doi: 10.1161/01.CIR.0000089507.19675.F9. [DOI] [PubMed] [Google Scholar]
  • 48.Moncada S, Higgs A. The L-arginine-nitric oxide pathway. N Engl J Med. 1993;329:2002–12. doi: 10.1056/NEJM199312303292706. [DOI] [PubMed] [Google Scholar]
  • 49.Luscher TF, Barton M. Biology of the endothelium. Clin Cardiol. 1997;20:II-3–10. [PubMed] [Google Scholar]
  • 50.Cernadas MR, Sanchez de Miguel L, Garcia-Duran M, Gonzalez-Fernandez F, Millas I, Monton M, et al. Expression of constitutive and inducible nitric oxide synthases in the vascular wall of young and aging rats. Circ Res. 1998;83:279–86. doi: 10.1161/01.res.83.3.279. [DOI] [PubMed] [Google Scholar]
  • 51.Chou TC, Yen MH, Li CY, Ding YA. Alterations of nitric oxide synthase expression with aging and hypertension in rats. Hypertension. 1998;31:643–8. doi: 10.1161/01.hyp.31.2.643. [DOI] [PubMed] [Google Scholar]
  • 52.Csiszar A, Labinskyy N, Smith K, Rivera A, Orosz Z, Ungvari Z. Vasculoprotective effects of anti-tumor necrosis factor-alpha treatment in aging. Am J Pathol. 2007;170:388–98. doi: 10.2353/ajpath.2007.060708. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.van der Loo B, Labugger R, Skepper JN, Bachschmid M, Kilo J, Powell JM, et al. Enhanced peroxynitrite formation is associated with vascular aging. J Exp Med. 2000;192:1731–44. doi: 10.1084/jem.192.12.1731. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Spier SA, Delp MD, Meininger CJ, Donato AJ, Ramsey MW, Muller-Delp JM. Effects of ageing and exercise training on endothelium-dependent vasodilatation and structure of rat skeletal muscle arterioles. J Physiol. 2004;556:947–58. doi: 10.1113/jphysiol.2003.060301. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Durrant JR, Seals DR, Connell ML, Russell MJ, Lawson BR, Folian BJ, et al. Voluntary wheel running restores endothelial function in conduit arteries of old mice: direct evidence for reduced oxidative stress, increased superoxide dismutase activity and down-regulation of NADPH oxidase. J Physiol. 2009;587:3271–85. doi: 10.1113/jphysiol.2009.169771. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Spier SA, Delp MD, Stallone JN, Dominguez JM, 2nd, Muller-Delp JM. Exercise training enhances flow-induced vasodilation in skeletal muscle resistance arteries of aged rats: role of PGI2 and nitric oxide. Am J Physiol Heart Circ Physiol. 2007;292:H3119–27. doi: 10.1152/ajpheart.00588.2006. [DOI] [PubMed] [Google Scholar]
  • 57.Nicholson WT, Vaa B, Hesse C, Eisenach JH, Joyner MJ. Aging is associated with reduced prostacyclin-mediated dilation in the human forearm. Hypertension. 2009;53:973–8. doi: 10.1161/HYPERTENSIONAHA.108.121483. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Novella S, Dantas AP, Segarra G, Novensa L, Heras M, Hermenegildo C, et al. Aging enhances contraction to thromboxane A2 in aorta from female senescence-accelerated mice. Age (Dordr) 2013;35:117–28. doi: 10.1007/s11357-011-9337-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Cardillo C, Kilcoyne CM, Cannon RO, 3rd, Panza JA. Interactions between nitric oxide and endothelin in the regulation of vascular tone of human resistance vessels in vivo. Hypertension. 2000;35:1237–41. doi: 10.1161/01.hyp.35.6.1237. [DOI] [PubMed] [Google Scholar]
  • 60.Tang EH, Vanhoutte PM. Gene expression changes of prostanoid synthases in endothelial cells and prostanoid receptors in vascular smooth muscle cells caused by aging and hypertension. Physiol Genomics. 2008;32:409–18. doi: 10.1152/physiolgenomics.00136.2007. [DOI] [PubMed] [Google Scholar]
  • 61.Goettsch W, Lattmann T, Amann K, Szibor M, Morawietz H, Munter K, et al. Increased expression of endothelin-1 and inducible nitric oxide synthase isoform II in aging arteries in vivo: implications for atherosclerosis. Biochem Biophys Res Commun. 2001;280:908–13. doi: 10.1006/bbrc.2000.4180. [DOI] [PubMed] [Google Scholar]
  • 62.Westby CM, Weil BR, Greiner JJ, Stauffer BL, DeSouza CA. Endothelin-1 vasoconstriction and the age-related decline in endothelium-dependent vasodilatation in men. Clin Sci (Lond) 2011;120:485–91. doi: 10.1042/CS20100475. [DOI] [PubMed] [Google Scholar]
  • 63.JI LL, LEEUWENBURGH C, LEICHTWEIS S, GORE M, FIEBIG R, HOLLANDER J, et al. Oxidative Stress and Aging: Role of Exercise and Its Influences on Antioxidant Systems. Ann NY Acad Sci. 1998;854:102–17. doi: 10.1111/j.1749-6632.1998.tb09896.x. [DOI] [PubMed] [Google Scholar]
  • 64.Moreau KL, Donato AJ, Tanaka H, Jones PP, Gates PE, Seals DR. Basal leg blood flow in healthy women is related to age and hormone replacement therapy status. J Physiol. 2003;547:309–16. doi: 10.1113/jphysiol.2002.032524. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Franzoni F, Ghiadoni L, Galetta F, Plantinga Y, Lubrano V, Huang Y, et al. Physical activity, plasma antioxidant capacity, and endothelium-dependent vasodilation in young and older men. American Journal of Hypertension. 2005;18:510. doi: 10.1016/j.amjhyper.2004.11.006. [DOI] [PubMed] [Google Scholar]
  • 66.Taddei S, Virdis A, Ghiadoni L, Salvetti G, Bernini G, Magagna A, et al. Age-Related Reduction of NO Availability and Oxidative Stress in Humans. Hypertension. 2001;38:274–9. doi: 10.1161/01.hyp.38.2.274. [DOI] [PubMed] [Google Scholar]
  • 67.Cohen RA, Plane F, Najibi S, Huk I, Malinski T, Garland CJ. Nitric oxide is the mediator of both endothelium-dependent relaxation and hyperpolarization of the rabbit carotid artery. Proc Natl Acad Sci U S A. 1997;94:4193–8. doi: 10.1073/pnas.94.8.4193. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Moncada S, Palmer RM, Higgs EA. Nitric oxide: physiology, pathophysiology, and pharmacology. Pharmacol Rev. 1991;43:109–42. [PubMed] [Google Scholar]
  • 69.Persson MG, Gustafsson LE, Wiklund NP, Hedqvist P, Moncada S. Endogenous nitric oxide as a modulator of rabbit skeletal muscle microcirculation in vivo. Br J Pharmacol. 1990;100:463–6. doi: 10.1111/j.1476-5381.1990.tb15829.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Griendling KK, Sorescu D, Ushio-Fukai M. NAD(P)H Oxidase: Role in Cardiovascular Biology and Disease. Circ Res. 2000;86:494–501. doi: 10.1161/01.res.86.5.494. [DOI] [PubMed] [Google Scholar]
  • 71.Harrison DG. Endothelial function and oxidant stress. Clin Cardiol. 1997;20:II-11–7. [PubMed] [Google Scholar]
  • 72.Price DT, Vita JA, Keaney JF., Jr Redox control of vascular nitric oxide bioavailability. Antioxid Redox Signal. 2000;2:919–35. doi: 10.1089/ars.2000.2.4-919. [DOI] [PubMed] [Google Scholar]
  • 73.Cai H, Harrison DG. Endothelial dysfunction in cardiovascular diseases: the role of oxidant stress. Circ Res. 2000;87:840–4. doi: 10.1161/01.res.87.10.840. [DOI] [PubMed] [Google Scholar]
  • 74.Mervaala EM, Cheng ZJ, Tikkanen I, Lapatto R, Nurminen K, Vapaatalo H, et al. Endothelial dysfunction and xanthine oxidoreductase activity in rats with human renin and angiotensinogen genes. Hypertension. 2001;37:414–8. doi: 10.1161/01.hyp.37.2.414. [DOI] [PubMed] [Google Scholar]
  • 75.Hamilton CA, Brosnan MJ, McIntyre M, Graham D, Dominiczak AF. Superoxide Excess in Hypertension and Aging: A Common Cause of Endothelial Dysfunction. Hypertension. 2001;37:529–34. doi: 10.1161/01.hyp.37.2.529. [DOI] [PubMed] [Google Scholar]
  • 76.Dikalov SI, Ungvari Z. Role of mitochondrial oxidative stress in hypertension. Am J Physiol Heart Circ Physiol. 2013;305:H1417–27. doi: 10.1152/ajpheart.00089.2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Quijano C, Castro L, Peluffo G, Valez V, Radi R. Enhanced mitochondrial superoxide in hyperglycemic endothelial cells: direct measurements and formation of hydrogen peroxide and peroxynitrite. Am J Physiol Heart Circ Physiol. 2007;293:H3404–14. doi: 10.1152/ajpheart.00761.2007. [DOI] [PubMed] [Google Scholar]
  • 78.Baker CS, Hall RJ, Evans TJ, Pomerance A, Maclouf J, Creminon C, et al. Cyclooxygenase-2 is widely expressed in atherosclerotic lesions affecting native and transplanted human coronary arteries and colocalizes with inducible nitric oxide synthase and nitrotyrosine particularly in macrophages. Arterioscler Thromb Vasc Biol. 1999;19:646–55. doi: 10.1161/01.atv.19.3.646. [DOI] [PubMed] [Google Scholar]
  • 79.Myatt L, Rosenfield RB, Eis AL, Brockman DE, Greer I, Lyall F. Nitrotyrosine residues in placenta. Evidence of peroxynitrite formation and action. Hypertension. 1996;28:488–93. doi: 10.1161/01.hyp.28.3.488. [DOI] [PubMed] [Google Scholar]
  • 80.Miller SJ, Watson WC, Kerr KA, Labarrere CA, Chen NX, Deeg MA, et al. Development of progressive aortic vasculopathy in a rat model of aging. Am J Physiol Heart Circ Physiol. 2007;293:H2634–43. doi: 10.1152/ajpheart.00397.2007. [DOI] [PubMed] [Google Scholar]
  • 81.Oelze M, Kroller-Schon S, Steven S, Lubos E, Doppler C, Hausding M, et al. Glutathione peroxidase-1 deficiency potentiates dysregulatory modifications of endothelial nitric oxide synthase and vascular dysfunction in aging. Hypertension. 2014;63:390–6. doi: 10.1161/HYPERTENSIONAHA.113.01602. [DOI] [PubMed] [Google Scholar]
  • 82.Gong X, Ma Y, Ruan Y, Fu G, Wu S. Long-term atorvastatin improves age-related endothelial dysfunction by ameliorating oxidative stress and normalizing eNOS/iNOS imbalance in rat aorta. Exp Gerontol. 2014;52:9–17. doi: 10.1016/j.exger.2014.01.015. [DOI] [PubMed] [Google Scholar]
  • 83.Ungvari Z, Bailey-Downs L, Gautam T, Sosnowska D, Wang M, Monticone RE, et al. Age-associated vascular oxidative stress, Nrf2 dysfunction, and NF-{kappa}B activation in the nonhuman primate Macaca mulatta. J Gerontol A Biol Sci Med Sci. 2011;66:866–75. doi: 10.1093/gerona/glr092. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Uddin M, Yang H, Shi M, Polley-Mandal M, Guo Z. Elevation of oxidative stress in the aorta of genetically hypertensive mice. Mech Ageing Dev. 2003;124:811–7. doi: 10.1016/s0047-6374(03)00135-0. [DOI] [PubMed] [Google Scholar]
  • 85.Tatchum-Talom R, Martin DS. Tempol improves vascular function in the mesenteric vascular bed of senescent rats. Can J Physiol Pharmacol. 2004;82:200–7. doi: 10.1139/y04-010. [DOI] [PubMed] [Google Scholar]
  • 86.Trott DW, Seawright JW, Luttrell MJ, Woodman CR. NAD(P)H oxidase-derived reactive oxygen species contribute to age-related impairments of endothelium-dependent dilation in rat soleus feed arteries. J Appl Physiol (1985) 2011;110:1171–80. doi: 10.1152/japplphysiol.01037.2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Wever RM, van Dam T, van Rijn HJ, de Groot F, Rabelink TJ. Tetrahydrobiopterin regulates superoxide and nitric oxide generation by recombinant endothelial nitric oxide synthase. Biochem Biophys Res Commun. 1997;237:340–4. doi: 10.1006/bbrc.1997.7069. [DOI] [PubMed] [Google Scholar]
  • 88.Eskurza I, Myerburgh LA, Kahn ZD, Seals DR. Tetrahydrobiopterin augments endothelium-dependent dilatation in sedentary but not in habitually exercising older adults. J Physiol. 2005;568:1057–65. doi: 10.1113/jphysiol.2005.092734. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Sindler AL, Delp MD, Reyes R, Wu G, Muller-Delp JM. Effects of ageing and exercise training on eNOS uncoupling in skeletal muscle resistance arterioles. J Physiol. 2009;587:3885–97. doi: 10.1113/jphysiol.2009.172221. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Eskurza I, Kahn Z, Seals D. Xanthine oxidase does not contribute to impaired peripheral artery conduit artery endothelium-dependent dilatation with aging. J Physiol. 2006;571:661–8. doi: 10.1113/jphysiol.2005.102566. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Donato AJ, Black AD, Jablonski KL, Gano LB, Seals DR. Aging is associated with greater nuclear NFkappaB, reduced IkappaBalpha, and increased expression of proinflammatory cytokines in vascular endothelial cells of healthy humans. Aging Cell. 2008;7:805–12. doi: 10.1111/j.1474-9726.2008.00438.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Donato AJ, Eskurza I, Jablonski KL, Gano LB, Pierce GL, Seals DR. Cytochrome P-450 2C9 signaling does not contribute to age-associated vascular endothelial dysfunction in humans. J Appl Physiol. 2008;105:1359–63. doi: 10.1152/japplphysiol.90629.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Ungvari ZI, Labinskyy N, Gupte SA, Chander PN, Edwards JG, Csiszar A. Dysregulation of mitochondrial biogenesis in vascular endothelial and smooth muscle cells of aged rats. Am J Physiol Heart Circ Physiol. 2008 doi: 10.1152/ajpheart.00012.2008. [DOI] [PubMed] [Google Scholar]
  • 94.Gioscia-Ryan RA, LaRocca TJ, Sindler AL, Zigler MC, Murphy MP, Seals DR. Mitochondria-targeted antioxidant (MitoQ) ameliorates age-related arterial endothelial dysfunction in mice. J Physiol. 2014;592:2549–61. doi: 10.1113/jphysiol.2013.268680. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Francia P, delli Gatti C, Bachschmid M, Martin-Padura I, Savoia C, Migliaccio E, et al. Deletion of p66shc gene protects against age-related endothelial dysfunction. Circulation. 2004;110:2889–95. doi: 10.1161/01.CIR.0000147731.24444.4D. [DOI] [PubMed] [Google Scholar]
  • 96.Brown KA, Didion SP, Andresen JJ, Faraci FM. Effect of aging, MnSOD deficiency, and genetic background on endothelial function: evidence for MnSOD haploinsufficiency. Arterioscler Thromb Vasc Biol. 2007;27:1941–6. doi: 10.1161/ATVBAHA.107.146852. [DOI] [PubMed] [Google Scholar]
  • 97.Dikalov SI, Dikalova AE, Bikineyeva AT, Schmidt HH, Harrison DG, Griendling KK. Distinct roles of Nox1 and Nox4 in basal and angiotensin II-stimulated superoxide and hydrogen peroxide production. Free Radic Biol Med. 2008;45:1340–51. doi: 10.1016/j.freeradbiomed.2008.08.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Woodman CR, Price EM, Laughlin MH. Aging induces muscle-specific impairment of endothelium-dependent dilation in skeletal muscle feed arteries. J Appl Physiol. 2002;93:1685–90. doi: 10.1152/japplphysiol.00461.2002. [DOI] [PubMed] [Google Scholar]
  • 99.Tian L, Cai Q, Wei H. Alterations of antioxidant enzymes and oxidative damage to macromolecules in different organs of rats during aging. Free Radic Biol Med. 1998;24:1477–84. doi: 10.1016/s0891-5849(98)00025-2. [DOI] [PubMed] [Google Scholar]
  • 100.Demaree SR, Lawler JM, Linehan J, Delp MD. Ageing alters aortic antioxidant enzyme activities in Fischer-344 rats. Acta Physiol Scand. 1999;166:203–8. doi: 10.1046/j.1365-201x.1999.00552.x. [DOI] [PubMed] [Google Scholar]
  • 101.Brandes RP, Fleming I, Busse R. Endothelial aging. Cardiovasc Res. 2005;66:286–94. doi: 10.1016/j.cardiores.2004.12.027. [DOI] [PubMed] [Google Scholar]
  • 102.Sindler AL, Reyes R, Chen B, Ghosh P, Gurovich AN, Kang LS, et al. Age and exercise training alter signaling through reactive oxygen species in the endothelium of skeletal muscle arterioles. J Appl Physiol (1985) 2013;114:681–93. doi: 10.1152/japplphysiol.00341.2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.MacMillan-Crow LA, Crow JP, Thompson JA. Peroxynitrite-mediated inactivation of manganese superoxide dismutase involves nitration and oxidation of critical tyrosine residues. Biochemistry. 1998;37:1613–22. doi: 10.1021/bi971894b. [DOI] [PubMed] [Google Scholar]
  • 104.Donato AJ, Walker AE, Magerko KA, Bramwell RC, Black AD, Henson GD, et al. Life-long caloric restriction reduces oxidative stress and preserves nitric oxide bioavailability and function in arteries of old mice. Aging Cell. 2013;12:772–83. doi: 10.1111/acel.12103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Walker AE, Henson GD, Reihl KD, Nielson EI, Morgan RG, Lesniewski LA, et al. Beneficial effects of lifelong caloric restriction on endothelial function are greater in conduit arteries compared to cerebral resistance arteries. Age (Dordr) 2014;36:559–69. doi: 10.1007/s11357-013-9585-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Donato AJ, Uberoi A, Bailey DM, Wray DW, Richardson RS. Exercise-induced brachial artery vasodilation: effects of antioxidants and exercise training in elderly men. Am J Physiol Heart Circ Physiol. 2010;298:H671–8. doi: 10.1152/ajpheart.00761.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Miura H, Bosnjak JJ, Ning G, Saito T, Miura M, Gutterman DD. Role for hydrogen peroxide in flow-induced dilation of human coronary arterioles. Circ Res. 2003;92:e31–40. doi: 10.1161/01.res.0000054200.44505.ab. [DOI] [PubMed] [Google Scholar]
  • 108.Kang LS, Reyes RA, Muller-Delp JM. Aging impairs flow-induced dilation in coronary arterioles: role of NO and H(2)O(2) Am J Physiol Heart Circ Physiol. 2009;297:H1087–95. doi: 10.1152/ajpheart.00356.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Krabbe K, Pedersen M, Bruunsgaard H. Inflammatory mediators in the elderly. Exp Gerontol. 2004;39:687–99. doi: 10.1016/j.exger.2004.01.009. [DOI] [PubMed] [Google Scholar]
  • 110.Vgontzas AN, Zoumakis M, Bixler EO, Lin HM, Prolo P, Vela-Bueno A, et al. Impaired nighttime sleep in healthy old versus young adults is associated with elevated plasma interleukin-6 and cortisol levels: physiologic and therapeutic implications. J Clin Endocrinol Metab. 2003;88:2087–95. doi: 10.1210/jc.2002-021176. [DOI] [PubMed] [Google Scholar]
  • 111.Belmin J, Bernard C, Corman B, Merval R, Esposito B, Tedgui A. Increased production of tumor necrosis factor and interleukin-6 by arterial wall of aged rats. Am J Physiol. 1995;268:H2288–93. doi: 10.1152/ajpheart.1995.268.6.H2288. [DOI] [PubMed] [Google Scholar]
  • 112.Lesniewski LA, Durrant JR, Connell ML, Henson GD, Black AD, Donato AJ, et al. Aerobic exercise reverses arterial inflammation with aging in mice. Am J Physiol Heart Circ Physiol. 2011;301:H1025–32. doi: 10.1152/ajpheart.01276.2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Lesniewski LA, Durrant JR, Connell ML, Folian BJ, Donato AJ, Seals DR. Salicylate treatment improves age-associated vascular endothelial dysfunction: potential role of nuclear factor kappaB and forkhead Box O phosphorylation. J Gerontol A Biol Sci Med Sci. 2011;66:409–18. doi: 10.1093/gerona/glq233. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Wang M, Zhang J, Jiang LQ, Spinetti G, Pintus G, Monticone R, et al. Proinflammatory profile within the grossly normal aged human aortic wall. Hypertension. 2007;50:219–27. doi: 10.1161/HYPERTENSIONAHA.107.089409. [DOI] [PubMed] [Google Scholar]
  • 115.Read MA, Whitley MZ, Williams AJ, Collins T. NF-kappa B and I kappa B alpha: an inducible regulatory system in endothelial activation. J Exp Med. 1994;179:503–12. doi: 10.1084/jem.179.2.503. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Karin M, Delhase M. The I kappa B kinase (IKK) and NF-kappa B: key elements of proinflammatory signalling. Semin Immunol. 2000;12:85–98. doi: 10.1006/smim.2000.0210. [DOI] [PubMed] [Google Scholar]
  • 117.van den Berg R, Haenen GR, van den Berg H, Bast A. Transcription factor NF-kappaB as a potential biomarker for oxidative stress. Br J Nutr. 2001;86 (Suppl 1):S121–7. doi: 10.1079/bjn2001340. [DOI] [PubMed] [Google Scholar]
  • 118.Schreck R, Albermann K, Baeuerle PA. Nuclear factor kappa B: an oxidative stress-responsive transcription factor of eukaryotic cells (a review) Free Radic Res Commun. 1992;17:221–37. doi: 10.3109/10715769209079515. [DOI] [PubMed] [Google Scholar]
  • 119.Schreck R, Rieber P, Baeuerle PA. Reactive oxygen intermediates as apparently widely used messengers in the activation of the NF-kappa B transcription factor and HIV-1. Embo J. 1991;10:2247–58. doi: 10.1002/j.1460-2075.1991.tb07761.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Karin M, Ben-Neriah Y. Phosphorylation meets ubiquitination: the control of NF-[kappa]B activity. Annu Rev Immunol. 2000;18:621–63. doi: 10.1146/annurev.immunol.18.1.621. [DOI] [PubMed] [Google Scholar]
  • 121.Walker AE, Kaplon RE, Pierce GL, Nowlan MJ, Seals DR. Prevention of age-related endothelial dysfunction by habitual aerobic exercise in healthy humans: possible role of nuclear factor kappaB. Clin Sci (Lond) 2014;127:645–54. doi: 10.1042/CS20140030. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Pierce GL, Lesniewski LA, Lawson BR, Beske SD, Seals DR. Nuclear factor-{kappa}B activation contributes to vascular endothelial dysfunction via oxidative stress in overweight/obese middle-aged and older humans. Circulation. 2009;119:1284–92. doi: 10.1161/CIRCULATIONAHA.108.804294. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Csiszar A, Smith K, Labinskyy N, Orosz Z, Rivera A, Ungvari Z. Resveratrol attenuates TNF-alpha-induced activation of coronary arterial endothelial cells: role of NF-kappaB inhibition. Am J Physiol Heart Circ Physiol. 2006;291:H1694–9. doi: 10.1152/ajpheart.00340.2006. [DOI] [PubMed] [Google Scholar]
  • 124.Xiao J, Song J, Hodara V, Ford A, Wang XL, Shi Q, et al. Protective Effects of Resveratrol on TNF-alpha-Induced Endothelial Cytotoxicity in Baboon Femoral Arterial Endothelial Cells. J Diabetes Res. 2013;2013:185172. doi: 10.1155/2013/185172. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Donato AJ, Henson GD, Morgan RG, Enz RA, Walker AE, Lesniewski LA. TNF-alpha impairs endothelial function in adipose tissue resistance arteries of mice with diet-induced obesity. Am J Physiol Heart Circ Physiol. 2012;303:H672–9. doi: 10.1152/ajpheart.00271.2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Manea A, Manea SA, Gafencu AV, Raicu M. Regulation of NADPH oxidase subunit p22(phox) by NF-kB in human aortic smooth muscle cells. Arch Physiol Biochem. 2007;113:163–72. doi: 10.1080/13813450701531235. [DOI] [PubMed] [Google Scholar]
  • 127.Kuwano Y, Kawahara T, Yamamoto H, Teshima-Kondo S, Tominaga K, Masuda K, et al. Interferon-gamma activates transcription of NADPH oxidase 1 gene and upregulates production of superoxide anion by human large intestinal epithelial cells. Am J Physiol Cell Physiol. 2006;290:C433–43. doi: 10.1152/ajpcell.00135.2005. [DOI] [PubMed] [Google Scholar]
  • 128.Anrather J, Racchumi G, Iadecola C. NF-kappaB regulates phagocytic NADPH oxidase by inducing the expression of gp91phox. J Biol Chem. 2006;281:5657–67. doi: 10.1074/jbc.M506172200. [DOI] [PubMed] [Google Scholar]
  • 129.Hasegawa Y, Saito T, Ogihara T, Ishigaki Y, Yamada T, Imai J, et al. Blockade of the nuclear factor-kappaB pathway in the endothelium prevents insulin resistance and prolongs life spans. Circulation. 2012;125:1122–33. doi: 10.1161/CIRCULATIONAHA.111.054346. [DOI] [PubMed] [Google Scholar]
  • 130.Schraufstatter IU, Chung J, Burger M. IL-8 activates endothelial cell CXCR1 and CXCR2 through Rho and Rac signaling pathways. Am J Physiol Lung Cell Mol Physiol. 2001;280:L1094–103. doi: 10.1152/ajplung.2001.280.6.L1094. [DOI] [PubMed] [Google Scholar]
  • 131.Mannell HK, Pircher J, Chaudhry DI, Alig SK, Koch EG, Mettler R, et al. ARNO regulates VEGF-dependent tissue responses by stabilizing endothelial VEGFR-2 surface expression. Cardiovasc Res. 2012;93:111–9. doi: 10.1093/cvr/cvr265. [DOI] [PubMed] [Google Scholar]
  • 132.Zhu W, London NR, Gibson CC, Davis CT, Tong Z, Sorensen LK, et al. Interleukin receptor activates a MYD88-ARNO-ARF6 cascade to disrupt vascular stability. Nature. 2012;492:252–5. doi: 10.1038/nature11603. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.van Wetering S, van den Berk N, van Buul JD, Mul FP, Lommerse I, Mous R, et al. VCAM-1-mediated Rac signaling controls endothelial cell-cell contacts and leukocyte transmigration. Am J Physiol Cell Physiol. 2003;285:C343–52. doi: 10.1152/ajpcell.00048.2003. [DOI] [PubMed] [Google Scholar]
  • 134.Jones CA, Nishiya N, London NR, Zhu W, Sorensen LK, Chan AC, et al. Slit2-Robo4 signalling promotes vascular stability by blocking Arf6 activity. Nature cell biology. 2009;11:1325–31. doi: 10.1038/ncb1976. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Liu ZJ, Tan Y, Beecham GW, Seo DM, Tian R, Li Y, et al. Notch activation induces endothelial cell senescence and pro-inflammatory response: implication of Notch signaling in atherosclerosis. Atherosclerosis. 2012;225:296–303. doi: 10.1016/j.atherosclerosis.2012.04.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Venkatesh D, Fredette N, Rostama B, Tang Y, Vary CP, Liaw L, et al. RhoA-mediated signaling in Notch-induced senescence-like growth arrest and endothelial barrier dysfunction. Arterioscler Thromb Vasc Biol. 2011;31:876–82. doi: 10.1161/ATVBAHA.110.221945. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.Scaffidi P, Misteli T. Lamin A-dependent misregulation of adult stem cells associated with accelerated ageing. Nature cell biology. 2008;10:452–9. doi: 10.1038/ncb1708. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Scaffidi P, Misteli T. Lamin A-dependent nuclear defects in human aging. Science. 2006;312:1059–63. doi: 10.1126/science.1127168. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Ragnauth CD, Warren DT, Liu Y, McNair R, Tajsic T, Figg N, et al. Prelamin A acts to accelerate smooth muscle cell senescence and is a novel biomarker of human vascular aging. Circulation. 2010;121:2200–10. doi: 10.1161/CIRCULATIONAHA.109.902056. [DOI] [PubMed] [Google Scholar]
  • 140.Olive M, Harten I, Mitchell R, Beers JK, Djabali K, Cao K, et al. Cardiovascular pathology in Hutchinson-Gilford progeria: correlation with the vascular pathology of aging. Arterioscler Thromb Vasc Biol. 2010;30:2301–9. doi: 10.1161/ATVBAHA.110.209460. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Kovacic JC, Moreno P, Hachinski V, Nabel EG, Fuster V. Cellular senescence, vascular disease, and aging: Part 1 of a 2-part review. Circulation. 2011;123:1650–60. doi: 10.1161/CIRCULATIONAHA.110.007021. [DOI] [PubMed] [Google Scholar]
  • 142.Oakley R, Tharakan B. Vascular hyperpermeability and aging. Aging Dis. 2014;5:114–25. doi: 10.14336/AD.2014.0500114. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143.Krouwer VJ, Hekking LH, Langelaar-Makkinje M, Regan-Klapisz E, Post JA. Endothelial cell senescence is associated with disrupted cell-cell junctions and increased monolayer permeability. Vasc Cell. 2012;4:12. doi: 10.1186/2045-824X-4-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144.Coppé J-P, Desprez P-Y, Krtolica A, Campisi J. The Senescence-Associated Secretory Phenotype: The Dark Side of Tumor Suppression. Annual Review of Pathology: Mechanisms of Disease. 2010;5:99–118. doi: 10.1146/annurev-pathol-121808-102144. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Campisi J, d’Adda di Fagagna F. Cellular senescence: when bad things happen to good cells. Nature reviews Molecular cell biology. 2007;8:729–40. doi: 10.1038/nrm2233. [DOI] [PubMed] [Google Scholar]
  • 146.Hayflick L. The Limited in Vitro Lifetime of Human Diploid Cell Strains. Experimental cell research. 1965;37:614–36. doi: 10.1016/0014-4827(65)90211-9. [DOI] [PubMed] [Google Scholar]
  • 147.Marchand A, Atassi F, Gaaya A, Leprince P, Le Feuvre C, Soubrier F, et al. The Wnt/beta-catenin pathway is activated during advanced arterial aging in humans. Aging cell. 2011;10:220–32. doi: 10.1111/j.1474-9726.2010.00661.x. [DOI] [PubMed] [Google Scholar]
  • 148.Morgan RG, Ives SJ, Lesniewski LA, Cawthon RM, Andtbacka RH, Noyes RD, et al. Age-related telomere uncapping is associated with cellular senescence and inflammation independent of telomere shortening in human arteries. American journal of physiology Heart and circulatory physiology. 2013 doi: 10.1152/ajpheart.00197.2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149.Matthews C, Gorenne I, Scott S, Figg N, Kirkpatrick P, Ritchie A, et al. Vascular smooth muscle cells undergo telomere-based senescence in human atherosclerosis: effects of telomerase and oxidative stress. Circulation research. 2006;99:156–64. doi: 10.1161/01.RES.0000233315.38086.bc. [DOI] [PubMed] [Google Scholar]
  • 150.Cafueri G, Parodi F, Pistorio A, Bertolotto M, Ventura F, Gambini C, et al. Endothelial and smooth muscle cells from abdominal aortic aneurysm have increased oxidative stress and telomere attrition. PloS one. 2012;7:e35312. doi: 10.1371/journal.pone.0035312. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151.Baker DJ, Wijshake T, Tchkonia T, LeBrasseur NK, Childs BG, van de Sluis B, et al. Clearance of p16Ink4a-positive senescent cells delays ageing-associated disorders. Nature. 2011;479:232–6. doi: 10.1038/nature10600. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152.Serrano M, Lin AW, McCurrach ME, Beach D, Lowe SW. Oncogenic ras provokes premature cell senescence associated with accumulation of p53 and p16INK4a. Cell. 1997;88:593–602. doi: 10.1016/s0092-8674(00)81902-9. [DOI] [PubMed] [Google Scholar]
  • 153.Parrinello S, Samper E, Krtolica A, Goldstein J, Melov S, Campisi J. Oxygen sensitivity severely limits the replicative lifespan of murine fibroblasts. Nature cell biology. 2003;5:741–7. doi: 10.1038/ncb1024. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154.Samper E, Nicholls DG, Melov S. Mitochondrial oxidative stress causes chromosomal instability of mouse embryonic fibroblasts. Aging cell. 2003;2:277–85. doi: 10.1046/j.1474-9728.2003.00062.x. [DOI] [PubMed] [Google Scholar]
  • 155.Di Leonardo A, Linke SP, Clarkin K, Wahl GM. DNA damage triggers a prolonged p53-dependent G1 arrest and long-term induction of Cip1 in normal human fibroblasts. Genes Dev. 1994;8:2540–51. doi: 10.1101/gad.8.21.2540. [DOI] [PubMed] [Google Scholar]
  • 156.Campisi J. Senescent cells, tumor suppression, and organismal aging: good citizens, bad neighbors. Cell. 2005;120:513–22. doi: 10.1016/j.cell.2005.02.003. [DOI] [PubMed] [Google Scholar]
  • 157.Kemp CJ, Wheldon T, Balmain A. p53-deficient mice are extremely susceptible to radiation-induced tumorigenesis. Nature genetics. 1994;8:66–9. doi: 10.1038/ng0994-66. [DOI] [PubMed] [Google Scholar]
  • 158.Stein GH, Drullinger LF, Soulard A, Dulic V. Differential roles for cyclin-dependent kinase inhibitors p21 and p16 in the mechanisms of senescence and differentiation in human fibroblasts. Molecular and cellular biology. 1999;19:2109–17. doi: 10.1128/mcb.19.3.2109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 159.Beausejour CM, Krtolica A, Galimi F, Narita M, Lowe SW, Yaswen P, et al. Reversal of human cellular senescence: roles of the p53 and p16 pathways. The EMBO journal. 2003;22:4212–22. doi: 10.1093/emboj/cdg417. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 160.Jacobs JJ, de Lange T. Significant role for p16INK4a in p53-independent telomere-directed senescence. Curr Biol. 2004;14:2302–8. doi: 10.1016/j.cub.2004.12.025. [DOI] [PubMed] [Google Scholar]
  • 161.Brookes S, Rowe J, Ruas M, Llanos S, Clark PA, Lomax M, et al. INK4a-deficient human diploid fibroblasts are resistant to RAS-induced senescence. The EMBO journal. 2002;21:2936–45. doi: 10.1093/emboj/cdf289. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 162.Macip S, Igarashi M, Fang L, Chen A, Pan ZQ, Lee SW, et al. Inhibition of p21-mediated ROS accumulation can rescue p21-induced senescence. The EMBO journal. 2002;21:2180–8. doi: 10.1093/emboj/21.9.2180. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163.Narita M, Nunez S, Heard E, Narita M, Lin AW, Hearn SA, et al. Rb-mediated heterochromatin formation and silencing of E2F target genes during cellular senescence. Cell. 2003;113:703–16. doi: 10.1016/s0092-8674(03)00401-x. [DOI] [PubMed] [Google Scholar]
  • 164.Coppe JP, Rodier F, Patil CK, Freund A, Desprez PY, Campisi J. Tumor suppressor and aging biomarker p16(INK4a) induces cellular senescence without the associated inflammatory secretory phenotype. The Journal of biological chemistry. 2011;286:36396–403. doi: 10.1074/jbc.M111.257071. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 165.Coppe JP, Patil CK, Rodier F, Sun Y, Munoz DP, Goldstein J, et al. Senescence-associated secretory phenotypes reveal cell-nonautonomous functions of oncogenic RAS and the p53 tumor suppressor. PLoS biology. 2008;6:2853–68. doi: 10.1371/journal.pbio.0060301. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166.Shelton DN, Chang E, Whittier PS, Choi D, Funk WD. Microarray analysis of replicative senescence. Curr Biol. 1999;9:939–45. doi: 10.1016/s0960-9822(99)80420-5. [DOI] [PubMed] [Google Scholar]
  • 167.Kyng KJ, May A, Stevnsner T, Becker KG, Kolvra S, Bohr VA. Gene expression responses to DNA damage are altered in human aging and in Werner Syndrome. Oncogene. 2005;24:5026–42. doi: 10.1038/sj.onc.1208692. [DOI] [PubMed] [Google Scholar]
  • 168.Yoon IK, Kim HK, Kim YK, Song IH, Kim W, Kim S, et al. Exploration of replicative senescence-associated genes in human dermal fibroblasts by cDNA microarray technology. Exp Gerontol. 2004;39:1369–78. doi: 10.1016/j.exger.2004.07.002. [DOI] [PubMed] [Google Scholar]
  • 169.Zhang H, Pan KH, Cohen SN. Senescence-specific gene expression fingerprints reveal cell-type-dependent physical clustering of up-regulated chromosomal loci. Proc Natl Acad Sci U S A. 2003;100:3251–6. doi: 10.1073/pnas.2627983100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 170.Maier JA, Voulalas P, Roeder D, Maciag T. Extension of the life-span of human endothelial cells by an interleukin-1 alpha antisense oligomer. Science. 1990;249:1570–4. doi: 10.1126/science.2218499. [DOI] [PubMed] [Google Scholar]
  • 171.Noureddine H, Gary-Bobo G, Alifano M, Marcos E, Saker M, Vienney N, et al. Pulmonary artery smooth muscle cell senescence is a pathogenic mechanism for pulmonary hypertension in chronic lung disease. Circulation research. 2011;109:543–53. doi: 10.1161/CIRCRESAHA.111.241299. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 172.Xin MG, Zhang J, Block ER, Patel JM. Senescence-enhanced oxidative stress is associated with deficiency of mitochondrial cytochrome c oxidase in vascular endothelial cells. Mechanisms of ageing and development. 2003;124:911–9. doi: 10.1016/s0047-6374(03)00163-5. [DOI] [PubMed] [Google Scholar]
  • 173.Sato I, Morita I, Kaji K, Ikeda M, Nagao M, Murota S. Reduction of nitric oxide producing activity associated with in vitro aging in cultured human umbilical vein endothelial cell. Biochemical and biophysical research communications. 1993;195:1070–6. doi: 10.1006/bbrc.1993.2153. [DOI] [PubMed] [Google Scholar]
  • 174.Minamino T, Miyauchi H, Yoshida T, Ishida Y, Yoshida H, Komuro I. Endothelial cell senescence in human atherosclerosis: role of telomere in endothelial dysfunction. Circulation. 2002;105:1541–4. doi: 10.1161/01.cir.0000013836.85741.17. [DOI] [PubMed] [Google Scholar]
  • 175.Chien Y, Scuoppo C, Wang X, Fang X, Balgley B, Bolden JE, et al. Control of the senescence-associated secretory phenotype by NF-kappaB promotes senescence and enhances chemosensitivity. Genes & development. 2011;25:2125–36. doi: 10.1101/gad.17276711. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 176.Salminen A, Kauppinen A, Kaarniranta K. Emerging role of NF-kappaB signaling in the induction of senescence-associated secretory phenotype (SASP) Cellular signalling. 2012;24:835–45. doi: 10.1016/j.cellsig.2011.12.006. [DOI] [PubMed] [Google Scholar]
  • 177.Tilstra JS, Robinson AR, Wang J, Gregg SQ, Clauson CL, Reay DP, et al. NF-kappaB inhibition delays DNA damage-induced senescence and aging in mice. The Journal of clinical investigation. 2012;122:2601–12. doi: 10.1172/JCI45785. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 178.Matoba S, Kang JG, Patino WD, Wragg A, Boehm M, Gavrilova O, et al. p53 regulates mitochondrial respiration. Science. 2006;312:1650–3. doi: 10.1126/science.1126863. [DOI] [PubMed] [Google Scholar]
  • 179.Mihm MJ, Jing L, Bauer JA. Nitrotyrosine causes selective vascular endothelial dysfunction and DNA damage. J Cardiovasc Pharmacol. 2000;36:182–7. doi: 10.1097/00005344-200008000-00007. [DOI] [PubMed] [Google Scholar]
  • 180.Ballinger SW, Patterson C, Yan CN, Doan R, Burow DL, Young CG, et al. Hydrogen peroxide- and peroxynitrite-induced mitochondrial DNA damage and dysfunction in vascular endothelial and smooth muscle cells. Circulation research. 2000;86:960–6. doi: 10.1161/01.res.86.9.960. [DOI] [PubMed] [Google Scholar]
  • 181.Mayr M, Hu Y, Hainaut H, Xu Q. Mechanical stress-induced DNA damage and rac-p38MAPK signal pathways mediate p53-dependent apoptosis in vascular smooth muscle cells. FASEB journal: official publication of the Federation of American Societies for Experimental Biology. 2002;16:1423–5. doi: 10.1096/fj.02-0042fje. [DOI] [PubMed] [Google Scholar]
  • 182.Singh NP, Muller CH, Berger RE. Effects of age on DNA double-strand breaks and apoptosis in human sperm. Fertility and sterility. 2003;80:1420–30. doi: 10.1016/j.fertnstert.2003.04.002. [DOI] [PubMed] [Google Scholar]
  • 183.Bua E, Johnson J, Herbst A, Delong B, McKenzie D, Salamat S, et al. Mitochondrial DNA-deletion mutations accumulate intracellularly to detrimental levels in aged human skeletal muscle fibers. American journal of human genetics. 2006;79:469–80. doi: 10.1086/507132. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 184.Herbst A, Pak JW, McKenzie D, Bua E, Bassiouni M, Aiken JM. Accumulation of mitochondrial DNA deletion mutations in aged muscle fibers: evidence for a causal role in muscle fiber loss. The journals of gerontology Series A, Biological sciences and medical sciences. 2007;62:235–45. doi: 10.1093/gerona/62.3.235. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 185.Aldred MA, Comhair SA, Varella-Garcia M, Asosingh K, Xu W, Noon GP, et al. Somatic chromosome abnormalities in the lungs of patients with pulmonary arterial hypertension. American journal of respiratory and critical care medicine. 2010;182:1153–60. doi: 10.1164/rccm.201003-0491OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 186.Spandidos DA, Ergazaki M, Arvanitis D, Kiaris H. Microsatellite instability in human atherosclerotic plaques. Biochemical and biophysical research communications. 1996;220:137–40. doi: 10.1006/bbrc.1996.0370. [DOI] [PubMed] [Google Scholar]
  • 187.Kiaris H, Hatzistamou J, Spandidos DA. Instability at the H-ras minisatellite in human atherosclerotic plaques. Atherosclerosis. 1996;125:47–51. doi: 10.1016/0021-9150(96)05841-8. [DOI] [PubMed] [Google Scholar]
  • 188.Inafuku M, Toda T, Iwasaki H, Oku H. Analysis of microsatellite instability and loss of heterozygosity in human aortic atherosclerotic lesions. Rinsho byori The Japanese journal of clinical pathology. 2004;52:961–5. [PubMed] [Google Scholar]
  • 189.Hatzistamou J, Kiaris H, Ergazaki M, Spandidos DA. Loss of heterozygosity and microsatellite instability in human atherosclerotic plaques. Biochemical and biophysical research communications. 1996;225:186–90. doi: 10.1006/bbrc.1996.1151. [DOI] [PubMed] [Google Scholar]
  • 190.Flouris GA, Arvanitis DA, Parissis JT, Arvanitis DL, Spandidos DA. Loss of heterozygosity in DNA mismatch repair genes in human atherosclerotic plaques. Molecular cell biology research communications: MCBRC. 2000;4:62–5. doi: 10.1006/mcbr.2000.0255. [DOI] [PubMed] [Google Scholar]
  • 191.Arvanitis DA, Flouris GA, Spandidos DA. Genomic rearrangements on VCAM1, SELE, APEG1and AIF1 loci in atherosclerosis. Journal of cellular and molecular medicine. 2005;9:153–9. doi: 10.1111/j.1582-4934.2005.tb00345.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 192.Durik M, Kavousi M, van der Pluijm I, Isaacs A, Cheng C, Verdonk K, et al. Nucleotide excision DNA repair is associated with age-related vascular dysfunction. Circulation. 2012;126:468–78. doi: 10.1161/CIRCULATIONAHA.112.104380. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 193.Harley CB, Futcher AB, Greider CW. Telomeres shorten during ageing of human fibroblasts. Nature. 1990;345:458–60. doi: 10.1038/345458a0. [DOI] [PubMed] [Google Scholar]
  • 194.Oikawa S, Tada-Oikawa S, Kawanishi S. Site-specific DNA damage at the GGG sequence by UVA involves acceleration of telomere shortening. Biochemistry. 2001;40:4763–8. doi: 10.1021/bi002721g. [DOI] [PubMed] [Google Scholar]
  • 195.Sitte N, Saretzki G, von Zglinicki T. Accelerated telomere shortening in fibroblasts after extended periods of confluency. Free radical biology & medicine. 1998;24:885–93. doi: 10.1016/s0891-5849(97)00363-8. [DOI] [PubMed] [Google Scholar]
  • 196.d’Adda di Fagagna F, Reaper PM, Clay-Farrace L, Fiegler H, Carr P, Von Zglinicki T, et al. A DNA damage checkpoint response in telomere-initiated senescence. Nature. 2003;426:194–8. doi: 10.1038/nature02118. [DOI] [PubMed] [Google Scholar]
  • 197.Takai H, Smogorzewska A, de Lange T. DNA damage foci at dysfunctional telomeres. Current biology: CB. 2003;13:1549–56. doi: 10.1016/s0960-9822(03)00542-6. [DOI] [PubMed] [Google Scholar]
  • 198.Kahn BB, Alquier T, Carling D, Hardie DG. AMP-activated protein kinase: ancient energy gauge provides clues to modern understanding of metabolism. Cell Metab. 2005;1:15–25. doi: 10.1016/j.cmet.2004.12.003. [DOI] [PubMed] [Google Scholar]
  • 199.Imai S, Armstrong CM, Kaeberlein M, Guarente L. Transcriptional silencing and longevity protein Sir2 is an NAD-dependent histone deacetylase. Nature. 2000;403:795–800. doi: 10.1038/35001622. [DOI] [PubMed] [Google Scholar]
  • 200.Tissenbaum HA, Guarente L. Increased dosage of a sir-2 gene extends lifespan in Caenorhabditis elegans. Nature. 2001;410:227–30. doi: 10.1038/35065638. [DOI] [PubMed] [Google Scholar]
  • 201.Rodgers JT, Lerin C, Gerhart-Hines Z, Puigserver P. Metabolic adaptations through the PGC-1 alpha and SIRT1 pathways. FEBS Lett. 2008;582:46–53. doi: 10.1016/j.febslet.2007.11.034. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 202.Whittle JR, Powell MJ, Popov VM, Shirley LA, Wang C, Pestell RG. Sirtuins, nuclear hormone receptor acetylation and transcriptional regulation. Trends Endocrinol Metab. 2007;18:356–64. doi: 10.1016/j.tem.2007.07.007. [DOI] [PubMed] [Google Scholar]
  • 203.Guarente L. Sirtuins as potential targets for metabolic syndrome. Nature. 2006;444:868–74. doi: 10.1038/nature05486. [DOI] [PubMed] [Google Scholar]
  • 204.Cohen HY, Miller C, Bitterman KJ, Wall NR, Hekking B, Kessler B, et al. Calorie restriction promotes mammalian cell survival by inducing the SIRT1 deacetylase. Science. 2004;305:390–2. doi: 10.1126/science.1099196. [DOI] [PubMed] [Google Scholar]
  • 205.Dali-Youcef N, Lagouge M, Froelich S, Koehl C, Schoonjans K, Auwerx J. Sirtuins: the ‘magnificent seven’, function, metabolism and longevity. Ann Med. 2007;39:335–45. doi: 10.1080/07853890701408194. [DOI] [PubMed] [Google Scholar]
  • 206.Kao CL, Chen LK, Chang YL, Yung MC, Hsu CC, Chen YC, et al. Resveratrol protects human endothelium from H(2)O(2)-induced oxidative stress and senescence via SirT1 activation. J Atheroscler Thromb. 2010;17:970–9. doi: 10.5551/jat.4333. [DOI] [PubMed] [Google Scholar]
  • 207.Donato AJ, Magerko KA, Lawson BR, Durrant JR, Lesniewski LA, Seals DR. SIRT-1 and vascular endothelial dysfunction with ageing in mice and humans. The Journal of physiology. 2011;589:4545–54. doi: 10.1113/jphysiol.2011.211219. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 208.Lafontaine-Lacasse M, Richard D, Picard F. Effects of age and gender on Sirt 1 mRNA expressions in the hypothalamus of the mouse. Neurosci Lett. 2010;480:1–3. doi: 10.1016/j.neulet.2010.01.008. [DOI] [PubMed] [Google Scholar]
  • 209.Yeung F, Hoberg JE, Ramsey CS, Keller MD, Jones DR, Frye RA, et al. Modulation of NF-kappaB-dependent transcription and cell survival by the SIRT1 deacetylase. EMBO J. 2004;23:2369–80. doi: 10.1038/sj.emboj.7600244. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 210.Mitchell SJ, Martin-Montalvo A, Mercken EM, Palacios HH, Ward TM, Abulwerdi G, et al. The SIRT1 activator SRT1720 extends lifespan and improves health of mice fed a standard diet. Cell Rep. 2014;6:836–43. doi: 10.1016/j.celrep.2014.01.031. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 211.Mattagajasingh I, Kim CS, Naqvi A, Yamamori T, Hoffman TA, Jung SB, et al. SIRT1 promotes endothelium-dependent vascular relaxation by activating endothelial nitric oxide synthase. Proc Natl Acad Sci U S A. 2007;104:14855–60. doi: 10.1073/pnas.0704329104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 212.Rippe C, Lesniewski L, Connell M, LaRocca T, Donato A, Seals D. Short-term calorie restriction reverses vascular endothelial dysfunction in old mice by increasing nitric oxide and reducing oxidative stress. Aging Cell. 2010;9:304–12. doi: 10.1111/j.1474-9726.2010.00557.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 213.Gano LB, Donato AJ, Hazma M, Hearon C, Sindler A, Seals DR. The SIRT1 Activator SRT1720 Reverses Vascular Endothelial Dysfunction, Excessive Superoxide Production and Inflammation with Aging in Mice. American Journal of Phsiology - Heart and Ciculatory Physiology. doi: 10.1152/ajpheart.00377.2014. In Revision. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 214.Csiszar A, Labinskyy N, Jimenez R, Pinto JT, Ballabh P, Losonczy G, et al. Anti-oxidative and anti-inflammatory vasoprotective effects of caloric restriction in aging: role of circulating factors and SIRT1. Mech Ageing Dev. 2009;130:518–27. doi: 10.1016/j.mad.2009.06.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 215.Das DK, Maulik N. Resveratrol in cardioprotection: a therapeutic promise of alternative medicine. Mol Interv. 2006;6:36–47. doi: 10.1124/mi.6.1.7. [DOI] [PubMed] [Google Scholar]
  • 216.Smith JJ, Kenney RD, Gagne DJ, Frushour BP, Ladd W, Galonek HL, et al. Small molecule activators of SIRT1 replicate signaling pathways triggered by calorie restriction in vivo. BMC Syst Biol. 2009;3:31. doi: 10.1186/1752-0509-3-31. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 217.Stivala LA, Savio M, Carafoli F, Perucca P, Bianchi L, Maga G, et al. Specific structural determinants are responsible for the antioxidant activity and the cell cycle effects of resveratrol. J Biol Chem. 2001;276:22586–94. doi: 10.1074/jbc.M101846200. [DOI] [PubMed] [Google Scholar]
  • 218.Hung LM, Chen JK, Huang SS, Lee RS, Su MJ. Cardioprotective effect of resveratrol, a natural antioxidant derived from grapes. Cardiovasc Res. 2000;47:549–55. doi: 10.1016/s0008-6363(00)00102-4. [DOI] [PubMed] [Google Scholar]
  • 219.Leiro J, Alvarez E, Arranz JA, Laguna R, Uriarte E, Orallo F. Effects of cis-resveratrol on inflammatory murine macrophages: antioxidant activity and down-regulation of inflammatory genes. J Leukoc Biol. 2004;75:1156–65. doi: 10.1189/jlb.1103561. [DOI] [PubMed] [Google Scholar]
  • 220.Labinskyy N, Csiszar A, Veress G, Stef G, Pacher P, Oroszi G, et al. Vascular dysfunction in aging: potential effects of resveratrol, an anti-inflammatory phytoestrogen. Curr Med Chem. 2006;13:989–96. doi: 10.2174/092986706776360987. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 221.Pervaiz S. Resveratrol: from grapevines to mammalian biology. Faseb J. 2003;17:1975–85. doi: 10.1096/fj.03-0168rev. [DOI] [PubMed] [Google Scholar]
  • 222.Pearson KJ, Baur JA, Lewis KN, Peshkin L, Price NL, Labinskyy N, et al. Resveratrol delays age-related deterioration and mimics transcriptional aspects of dietary restriction without extending life span. Cell Metab. 2008;8:157–68. doi: 10.1016/j.cmet.2008.06.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 223.Gano LB, Seals DR, Hazma M, Lesniewski LA, Donato AJ. Chronic Treatment with SIRT1 Activator SRT1720 Ameliorates Age-Associated Vascular Endothelial Dysfunction in B6D2F1 Mice. Circulation. 2010;122:Abstract A10813. [Google Scholar]
  • 224.Hattori Y, Suzuki K, Hattori S, Kasai K. Metformin inhibits cytokine-induced nuclear factor kappaB activation via AMP-activated protein kinase activation in vascular endothelial cells. Hypertension. 2006;47:1183–8. doi: 10.1161/01.HYP.0000221429.94591.72. [DOI] [PubMed] [Google Scholar]
  • 225.Hardie DG, Hawley SA, Scott JW. AMP-activated protein kinase--development of the energy sensor concept. J Physiol. 2006;574:7–15. doi: 10.1113/jphysiol.2006.108944. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 226.Fisslthaler B, Fleming I. Activation and signaling by the AMP-activated protein kinase in endothelial cells. Circ Res. 2009;105:114–27. doi: 10.1161/CIRCRESAHA.109.201590. [DOI] [PubMed] [Google Scholar]
  • 227.Sanz P. AMP-activated protein kinase: structure and regulation. Curr Protein Pept Sci. 2008;9:478–92. doi: 10.2174/138920308785915254. [DOI] [PubMed] [Google Scholar]
  • 228.Wong AK, Howie J, Petrie JR, Lang CC. AMP-activated protein kinase pathway: a potential therapeutic target in cardiometabolic disease. Clin Sci (Lond) 2009;116:607–20. doi: 10.1042/CS20080066. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 229.Owen MR, Doran E, Halestrap AP. Evidence that metformin exerts its anti-diabetic effects through inhibition of complex 1 of the mitochondrial respiratory chain. Biochem J. 2000;348(Pt 3):607–14. [PMC free article] [PubMed] [Google Scholar]
  • 230.Stephenne X, Foretz M, Taleux N, van der Zon GC, Sokal E, Hue L, et al. Metformin activates AMP-activated protein kinase in primary human hepatocytes by decreasing cellular energy status. Diabetologia. 2011;54:3101–10. doi: 10.1007/s00125-011-2311-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 231.Buzzai M, Jones RG, Amaravadi RK, Lum JJ, DeBerardinis RJ, Zhao F, et al. Systemic treatment with the antidiabetic drug metformin selectively impairs p53-deficient tumor cell growth. Cancer Res. 2007;67:6745–52. doi: 10.1158/0008-5472.CAN-06-4447. [DOI] [PubMed] [Google Scholar]
  • 232.Janjetovic K, Vucicevic L, Misirkic M, Vilimanovich U, Tovilovic G, Zogovic N, et al. Metformin reduces cisplatin-mediated apoptotic death of cancer cells through AMPK-independent activation of Akt. Eur J Pharmacol. 2011;651:41–50. doi: 10.1016/j.ejphar.2010.11.005. [DOI] [PubMed] [Google Scholar]
  • 233.Janjetovic K, Harhaji-Trajkovic L, Misirkic-Marjanovic M, Vucicevic L, Stevanovic D, Zogovic N, et al. In vitro and in vivo anti-melanoma action of metformin. Eur J Pharmacol. 2011;668:373–82. doi: 10.1016/j.ejphar.2011.07.004. [DOI] [PubMed] [Google Scholar]
  • 234.Saeedi R, Parsons HL, Wambolt RB, Paulson K, Sharma V, Dyck JR, et al. Metabolic actions of metformin in the heart can occur by AMPK-independent mechanisms. Am J Physiol Heart Circ Physiol. 2008;294:H2497–506. doi: 10.1152/ajpheart.00873.2007. [DOI] [PubMed] [Google Scholar]
  • 235.Li X, Han Y, Pang W, Li C, Xie X, Shyy JY, et al. AMP-activated protein kinase promotes the differentiation of endothelial progenitor cells. Arterioscler Thromb Vasc Biol. 2008;28:1789–95. doi: 10.1161/ATVBAHA.108.172452. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 236.Ahluwalia A, Tarnawski AS. Activation of the metabolic sensor-AMP activated protein kinase reverses impairment of angiogenesis in aging myocardial microvascular endothelial cells. Implications for the aging heart. J Physiol Pharmacol. 2011;62:583–7. [PubMed] [Google Scholar]
  • 237.Ouchi N, Shibata R, Walsh K. AMP-activated protein kinase signaling stimulates VEGF expression and angiogenesis in skeletal muscle. Circ Res. 2005;96:838–46. doi: 10.1161/01.RES.0000163633.10240.3b. [DOI] [PubMed] [Google Scholar]
  • 238.Nagata D, Mogi M, Walsh K. AMP-activated protein kinase (AMPK) signaling in endothelial cells is essential for angiogenesis in response to hypoxic stress. J Biol Chem. 2003;278:31000–6. doi: 10.1074/jbc.M300643200. [DOI] [PubMed] [Google Scholar]
  • 239.Kanazawa I, Yamaguchi T, Yano S, Yamauchi M, Sugimoto T. Metformin enhances the differentiation and mineralization of osteoblastic MC3T3-E1 cells via AMP kinase activation as well as eNOS and BMP-2 expression. Biochemical and biophysical research communications. 2008;375:414–9. doi: 10.1016/j.bbrc.2008.08.034. [DOI] [PubMed] [Google Scholar]
  • 240.Levine YC, Li GK, Michel T. Agonist-modulated regulation of AMP-activated protein kinase (AMPK) in endothelial cells. Evidence for an AMPK -> Rac1 -> Akt -> endothelial nitric-oxide synthase pathway. J Biol Chem. 2007;282:20351–64. doi: 10.1074/jbc.M702182200. [DOI] [PubMed] [Google Scholar]
  • 241.Chen ZP, Mitchelhill KI, Michell BJ, Stapleton D, Rodriguez-Crespo I, Witters LA, et al. AMP-activated protein kinase phosphorylation of endothelial NO synthase. FEBS Lett. 1999;443:285–9. doi: 10.1016/s0014-5793(98)01705-0. [DOI] [PubMed] [Google Scholar]
  • 242.Chen Z, Peng IC, Sun W, Su MI, Hsu PH, Fu Y, et al. AMP-activated protein kinase functionally phosphorylates endothelial nitric oxide synthase Ser633. Circ Res. 2009;104:496–505. doi: 10.1161/CIRCRESAHA.108.187567. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 243.Majithiya JB, Balaraman R. Metformin reduces blood pressure and restores endothelial function in aorta of streptozotocin-induced diabetic rats. Life Sci. 2006;78:2615–24. doi: 10.1016/j.lfs.2005.10.020. [DOI] [PubMed] [Google Scholar]
  • 244.Sartoretto JL, Melo GA, Carvalho MH, Nigro D, Passaglia RT, Scavone C, et al. Metformin treatment restores the altered microvascular reactivity in neonatal streptozotocin-induced diabetic rats increasing NOS activity, but not NOS expression. Life Sci. 2005;77:2676–89. doi: 10.1016/j.lfs.2005.05.022. [DOI] [PubMed] [Google Scholar]
  • 245.Katakam PV, Ujhelyi MR, Hoenig M, Miller AW. Metformin improves vascular function in insulin-resistant rats. Hypertension. 2000;35:108–12. doi: 10.1161/01.hyp.35.1.108. [DOI] [PubMed] [Google Scholar]
  • 246.Zu Y, Liu L, Lee MY, Xu C, Liang Y, Man RY, et al. SIRT1 promotes proliferation and prevents senescence through targeting LKB1 in primary porcine aortic endothelial cells. Circ Res. 2010;106:1384–93. doi: 10.1161/CIRCRESAHA.109.215483. [DOI] [PubMed] [Google Scholar]
  • 247.Lesniewski LA, Zigler MC, Durrant JR, Donato AJ, Seals DR. Sustained activation of AMPK ameliorates age-associated vascular endothelial dysfunction via a nitric oxide-independent mechanism. Mech Ageing Dev. 2012;133:368–71. doi: 10.1016/j.mad.2012.03.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 248.Pu Y, Zhang H, Wang P, Zhao Y, Li Q, Wei X, et al. Dietary curcumin ameliorates aging-related cerebrovascular dysfunction through the AMPK/uncoupling protein 2 pathway. Cell Physiol Biochem. 2013;32:1167–77. doi: 10.1159/000354516. [DOI] [PubMed] [Google Scholar]
  • 249.Davis B, Rahman A, Arner A. AMP-activated kinase relaxes agonist induced contractions in the mouse aorta via effects on PKC signaling and inhibits NO-induced relaxation. Eur J Pharmacol. 2012;695:88–95. doi: 10.1016/j.ejphar.2012.07.025. [DOI] [PubMed] [Google Scholar]
  • 250.Sharp ZD, Strong R. The role of mTOR signaling in controlling mammalian life span: what a fungicide teaches us about longevity. J Gerontol A Biol Sci Med Sci. 2010;65:580–9. doi: 10.1093/gerona/glp212. [DOI] [PubMed] [Google Scholar]
  • 251.Guertin DA, Sabatini DM. Defining the role of mTOR in cancer. Cancer Cell. 2007;12:9–22. doi: 10.1016/j.ccr.2007.05.008. [DOI] [PubMed] [Google Scholar]
  • 252.Vellai T, Takacs-Vellai K, Zhang Y, Kovacs AL, Orosz L, Muller F. Genetics: influence of TOR kinase on lifespan in C. elegans. Nature. 2003;426:620. doi: 10.1038/426620a. [DOI] [PubMed] [Google Scholar]
  • 253.Powers RW, 3rd, Kaeberlein M, Caldwell SD, Kennedy BK, Fields S. Extension of chronological life span in yeast by decreased TOR pathway signaling. Genes Dev. 2006;20:174–84. doi: 10.1101/gad.1381406. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 254.Kapahi P, Zid BM, Harper T, Koslover D, Sapin V, Benzer S. Regulation of lifespan in Drosophila by modulation of genes in the TOR signaling pathway. Current biology: CB. 2004;14:885–90. doi: 10.1016/j.cub.2004.03.059. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 255.Jia K, Chen D, Riddle DL. The TOR pathway interacts with the insulin signaling pathway to regulate C. elegans larval development, metabolism and life span. Development. 2004;131:3897–906. doi: 10.1242/dev.01255. [DOI] [PubMed] [Google Scholar]
  • 256.Harrison DE, Strong R, Sharp ZD, Nelson JF, Astle CM, Flurkey K, et al. Rapamycin fed late in life extends lifespan in genetically heterogeneous mice. Nature. 2009;460:392–5. doi: 10.1038/nature08221. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 257.Wilkinson JE, Burmeister L, Brooks SV, Chan CC, Friedline S, Harrison DE, et al. Rapamycin slows aging in mice. Aging Cell. 2012;11:675–82. doi: 10.1111/j.1474-9726.2012.00832.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 258.Selman C, Tullet JM, Wieser D, Irvine E, Lingard SJ, Choudhury AI, et al. Ribosomal protein S6 kinase 1 signaling regulates mammalian life span. Science. 2009;326:140–4. doi: 10.1126/science.1177221. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 259.Reihl K, Seals D, Henson G, LaRocca T, Magerko K, Bosshardt G, et al. Dietary rapamycin selectively improves arterial function in old mice. FASEB Journal. 2013;27:1194.17. [Google Scholar]
  • 260.Sethi G, Sung B, Aggarwal BB. Nuclear factor-kappaB activation: from bench to bedside. Exp Biol Med (Maywood) 2008;233:21–31. doi: 10.3181/0707-MR-196. [DOI] [PubMed] [Google Scholar]
  • 261.Tunon MJ, Sanchez-Campos S, Gutierrez B, Culebras JM, Gonzalez-Gallego J. Effects of FK506 and rapamycin on generation of reactive oxygen species, nitric oxide production and nuclear factor kappa B activation in rat hepatocytes. Biochem Pharmacol. 2003;66:439–45. doi: 10.1016/s0006-2952(03)00288-0. [DOI] [PubMed] [Google Scholar]
  • 262.Lai JH, Tan TH. CD28 signaling causes a sustained down-regulation of I kappa B alpha which can be prevented by the immunosuppressant rapamycin. J Biol Chem. 1994;269:30077–80. [PubMed] [Google Scholar]
  • 263.Romano MF, Avellino R, Petrella A, Bisogni R, Romano S, Venuta S. Rapamycin inhibits doxorubicin-induced NF-kappaB/Rel nuclear activity and enhances the apoptosis of melanoma cells. Eur J Cancer. 2004;40:2829–36. doi: 10.1016/j.ejca.2004.08.017. [DOI] [PubMed] [Google Scholar]
  • 264.Salminen A, Hyttinen JM, Kaarniranta K. AMP-activated protein kinase inhibits NF-kappaB signaling and inflammation: impact on healthspan and lifespan. J Mol Med (Berl) 2011;89:667–76. doi: 10.1007/s00109-011-0748-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 265.Yang Z, Kahn BB, Shi H, Xue BZ. Macrophage alpha1 AMP-activated protein kinase (alpha1AMPK) antagonizes fatty acid-induced inflammation through SIRT1. J Biol Chem. 2010;285:19051–9. doi: 10.1074/jbc.M110.123620. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 266.Cacicedo JM, Yagihashi N, Keaney JF, Jr, Ruderman NB, Ido Y. AMPK inhibits fatty acid-induced increases in NF-kappaB transactivation in cultured human umbilical vein endothelial cells. Biochem Biophys Res Commun. 2004;324:1204–9. doi: 10.1016/j.bbrc.2004.09.177. [DOI] [PubMed] [Google Scholar]
  • 267.Katerelos M, Mudge SJ, Stapleton D, Auwardt RB, Fraser SA, Chen CG, et al. 5-aminoimidazole-4-carboxamide ribonucleoside and AMP-activated protein kinase inhibit signalling through NF-kappaB. Immunol Cell Biol. 2010;88:754–60. doi: 10.1038/icb.2010.44. [DOI] [PubMed] [Google Scholar]
  • 268.Liu C, Liang B, Wang Q, Wu J, Zou MH. Activation of AMP-activated protein kinase alpha1 alleviates endothelial cell apoptosis by increasing the expression of anti-apoptotic proteins Bcl-2 and survivin. J Biol Chem. 2010;285:15346–55. doi: 10.1074/jbc.M110.102491. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  • 269.Papanicolaou KN, Izumiya Y, Walsh K. Forkhead transcription factors and cardiovascular biology. Circ Res. 2008;102:16–31. doi: 10.1161/CIRCRESAHA.107.164186. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 270.Sedding DG. FoxO transcription factors in oxidative stress response and ageing--a new fork on the way to longevity? Biol Chem. 2008;389:279–83. doi: 10.1515/BC.2008.033. [DOI] [PubMed] [Google Scholar]
  • 271.Tsai KL, Sun YJ, Huang CY, Yang JY, Hung MC, Hsiao CD. Crystal structure of the human FOXO3a-DBD/DNA complex suggests the effects of post-translational modification. Nucleic Acids Res. 2007;35:6984–94. doi: 10.1093/nar/gkm703. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 272.Huang H, Regan KM, Wang F, Wang D, Smith DI, van Deursen JM, et al. Skp2 inhibits FOXO1 in tumor suppression through ubiquitin-mediated degradation. Proc Natl Acad Sci U S A. 2005;102:1649–54. doi: 10.1073/pnas.0406789102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 273.Huang H, Tindall DJ. Dynamic FoxO transcription factors. J Cell Sci. 2007;120:2479–87. doi: 10.1242/jcs.001222. [DOI] [PubMed] [Google Scholar]
  • 274.Brunet A, Sweeney LB, Sturgill JF, Chua KF, Greer PL, Lin Y, et al. Stress-dependent regulation of FOXO transcription factors by the SIRT1 deacetylase. Science. 2004;303:2011–5. doi: 10.1126/science.1094637. [DOI] [PubMed] [Google Scholar]
  • 275.Paik JH. FOXOs in the maintenance of vascular homoeostasis. Biochem Soc Trans. 2006;34:731–4. doi: 10.1042/BST0340731. [DOI] [PubMed] [Google Scholar]
  • 276.Kobayashi Y, Furukawa-Hibi Y, Chen C, Horio Y, Isobe K, Ikeda K, et al. SIRT1 is critical regulator of FOXO-mediated transcription in response to oxidative stress. Int J Mol Med. 2005;16:237–43. [PubMed] [Google Scholar]
  • 277.Laplante M, Sabatini DM. mTOR signaling at a glance. J Cell Sci. 2009;122:3589–94. doi: 10.1242/jcs.051011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 278.Hong F, Larrea MD, Doughty C, Kwiatkowski DJ, Squillace R, Slingerland JM. mTOR-raptor binds and activates SGK1 to regulate p27 phosphorylation. Mol Cell. 2008;30:701–11. doi: 10.1016/j.molcel.2008.04.027. [DOI] [PubMed] [Google Scholar]
  • 279.Greer EL, Oskoui PR, Banko MR, Maniar JM, Gygi MP, Gygi SP, et al. The energy sensor AMP-activated protein kinase directly regulates the mammalian FOXO3 transcription factor. J Biol Chem. 2007;282:30107–19. doi: 10.1074/jbc.M705325200. [DOI] [PubMed] [Google Scholar]
  • 280.Ryan KM, Phillips AC, Vousden KH. Regulation and function of the p53 tumor suppressor protein. Curr Opin Cell Biol. 2001;13:332–7. doi: 10.1016/s0955-0674(00)00216-7. [DOI] [PubMed] [Google Scholar]
  • 281.Gorenne I, Kumar S, Gray K, Figg N, Yu H, Mercer J, et al. Vascular smooth muscle cell sirtuin 1 protects against DNA damage and inhibits atherosclerosis. Circulation. 2013;127:386–96. doi: 10.1161/CIRCULATIONAHA.112.124404. [DOI] [PubMed] [Google Scholar]
  • 282.Luo J, Nikolaev AY, Imai S, Chen D, Su F, Shiloh A, et al. Negative control of p53 by Sir2alpha promotes cell survival under stress. Cell. 2001;107:137–48. doi: 10.1016/s0092-8674(01)00524-4. [DOI] [PubMed] [Google Scholar]
  • 283.Vaziri H, Dessain SK, Ng Eaton E, Imai SI, Frye RA, Pandita TK, et al. hSIR2(SIRT1) functions as an NAD-dependent p53 deacetylase. Cell. 2001;107:149–59. doi: 10.1016/s0092-8674(01)00527-x. [DOI] [PubMed] [Google Scholar]
  • 284.Craig AL, Chrystal JA, Fraser JA, Sphyris N, Lin Y, Harrison BJ, et al. The MDM2 ubiquitination signal in the DNA-binding domain of p53 forms a docking site for calcium calmodulin kinase superfamily members. Mol Cell Biol. 2007;27:3542–55. doi: 10.1128/MCB.01595-06. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 285.Jones RG, Plas DR, Kubek S, Buzzai M, Mu J, Xu Y, et al. AMP-activated protein kinase induces a p53-dependent metabolic checkpoint. Mol Cell. 2005;18:283–93. doi: 10.1016/j.molcel.2005.03.027. [DOI] [PubMed] [Google Scholar]
  • 286.Thoreen CC, Sabatini DM. AMPK and p53 help cells through lean times. Cell Metab. 2005;1:287–8. doi: 10.1016/j.cmet.2005.04.009. [DOI] [PubMed] [Google Scholar]
  • 287.Feng Z, Zhang H, Levine AJ, Jin S. The coordinate regulation of the p53 and mTOR pathways in cells. Proc Natl Acad Sci U S A. 2005;102:8204–9. doi: 10.1073/pnas.0502857102. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES