Skip main navigation

Aging and Atherosclerosis

Mechanisms, Functional Consequences, and Potential Therapeutics for Cellular Senescence
Originally publishedhttps://doi.org/10.1161/CIRCRESAHA.111.261388Circulation Research. 2012;111:245–259

    Abstract

    Atherosclerosis is classed as a disease of aging, such that increasing age is an independent risk factor for the development of atherosclerosis. Atherosclerosis is also associated with premature biological aging, as atherosclerotic plaques show evidence of cellular senescence characterized by reduced cell proliferation, irreversible growth arrest and apoptosis, elevated DNA damage, epigenetic modifications, and telomere shortening and dysfunction. Not only is cellular senescence associated with atherosclerosis, there is growing evidence that cellular senescence promotes atherosclerosis. This review examines the pathology of normal vascular aging, the evidence for cellular senescence in atherosclerosis, the mechanisms underlying cellular senescence including reactive oxygen species, replication exhaustion and DNA damage, the functional consequences of vascular cell senescence, and the possibility that preventing accelerated cellular senescence is a therapeutic target in atherosclerosis.

    Introduction

    Atherosclerosis and the subsequent cardiovascular complications, such as myocardial infarction, stroke, and ischemic heart failure, is a major cause of death in the Western world. The risk factors of atherosclerosis are well known, including hypertension, diabetes, serum total and low-density lipoprotein (LDL) cholesterol, and smoking. Increasing evidence indicates that aging is also an important risk factor for atherosclerosis and persists as an independent contributor when all other known factors are controlled. Premature or accelerated vascular aging can be promoted by cardiovascular risk factors,1,2 and cellular senescence is also observed in patients with atherosclerosis.3 Atherosclerosis is therefore a disease of both organismal aging and cellular senescence par excellence.

    Pathology of the Advanced Atherosclerotic Plaque

    The mature atherosclerotic plaque comprises an accumulation of vascular smooth muscle cells (VSMCs) and their secreted products (collagen and elastin), inflammatory cells (macrophages, T lymphocytes, dendritic cells, and mast cells), and both intracellular and extracellular lipid and debris (Figure 1). The latter is often concentrated in a necrotic core surrounded by a fibrous cap composed predominately of VSMCs. The stability of the atherosclerotic plaque depends on the thickness of the fibrous cap and the degree of cap inflammation. Plaque rupture is increased by cap thinning, promoted by death of VSMCs and breakdown of collagen and extracellular matrix (ECM), which may subsequently lead to myocardial infarction or stroke. However, plaque rupture is frequently subclinical, as VSMCs repair the rupture and reorganize associated thrombus. Indeed, complicated plaques frequently show evidence of multiple ruptures and repair, ultimately resulting in luminal narrowing. Successful plaque repair requires VSMCs to proliferate and synthesize matrix, both properties that are altered by cellular senescence. Indeed, cellular senescence can alter many of the proatherogenic events seen in Figure 1.

    Figure 1.

    Figure 1. Schematic of atherogenesis and an unstable atherosclerotic plaque. The advanced atherosclerotic plaque consists of a fibrous cap rich in VSMCs and collagen, surrounding a “necrotic” core comprising lipids, foam cells, and debris. Plaque formation involves a series of events initiated from (1) EC dysfunction and activation with increased surface adhesion molecules on ECs, including ICAM-1 and VCAM-1, and augmented vascular permeability promoting the (2) attachment and infiltration of lipid and inflammatory cells into the subendothelial space. (3) Monocytes differentiate into macrophages, engulf lipid to form foam cells and release proinflammatory cytokines (eg, MCP1, IL-6, IL-1β) to create an inflammatory environment. (4) In early lesions, VSMCs proliferate/migrate in response to mitogens such as PDGF and synthesize collagen to form the fibrous cap that encloses the growing lipid core. At later stages, VSMCs undergo senescence and release multiple cytokines as part of the senescence-associated secretory phenotype (SASP), which augment the preexisting inflammation. (5) The gradual loss of VSMCs by apoptosis, in part mediated by engagement of death receptors on VSMCs with death ligands on macrophages and T lymphocytes, and increased activity of matrix-degrading enzymes in the cap result in (6) plaque rupture, with subsequent platelet attachment and thrombosis. Each of the processes involved in atherogenesis and plaque rupture indicated as (1) to (6) are promoted by cellular senescence (illustration: Cosmocyte/Ben Smith).

    Pathology of Normal and Premature Vascular Aging

    Vessel aging, even in the absence of atherosclerosis, leads to intimal and medial thickening (vascular remodeling) as well as gradual loss of arterial elasticity, resulting in vascular stiffness.3 Aged vessels show a number of characteristic pathological processes, many of which are also seen in atherosclerosis.4,5 For example, aged vessels show reduced medial VSMC number, increased collagen deposition, and fracture of the elastin lamellae, which may lead to vessel dilation and increased lumen size.6 Increased collagen and decreased elastin content, promoted at least in part by age-associated increases in glycated proteins, matrix metalloproteinase enzyme activity, and trophic stimuli such as angiotensin II signaling, impair vessel elasticity and hence promote vascular stiffness7 and subsequently hypertension.8 Hypertension can further stimulate collagen production with increased vessel stiffness and endothelial cell (EC) dysfunction. In addition to alterations in matrix and cell composition, aged vessels show elevated expression of a number of proinflammatory molecules9 and increased uptake of plasma lipoproteins.10 In part, these effects may be due to increased expression of leukocyte adhesion molecules on ECs in aged vessels, which trigger the familiar processes of monocyte migration followed by increased uptake of atherogenic lipoproteins with subsequent inflammation, key events that ultimately promote atherosclerosis (Figure 1). Aged ECs and VSMCs also show increased secretion of proinflammatory cytokines (see below), resulting in persistent vascular inflammation. Thus, the effects of atherosclerosis are superimposed on normal aging of the underlying vessel.

    A number of genetic diseases associated with premature aging also show the typical pathology of vascular aging and are prone to cardiovascular complications including atherosclerosis. For example, Hutchinson Gilford progeria syndrome (HGPS) is a rare, fatal, and progressive premature aging condition due to defects in lamin A that forms nuclear lamina. HGPS vessels from young patients reveal accumulated collagen, fractured elastin lamellae, and a thickened intima, with some vessels showing advanced atherosclerotic lesions containing chronic inflammation, calcification, and VSMC loss.11 Mouse models of HGPS recapitulate the vascular pathology in patients and also show increased vascular stiffness and hypertension, supporting the concept of premature vascular aging.12 Another example is Werner syndrome (WS), a loss-of-function mutation in the WS ATP-dependent helicase (WRN), which shows a similar pathology and accelerated atherosclerosis. WS knock-out mice also show insulin resistance and increased blood glucose, increased circulating cholesterol, and enhanced lipid deposition,13 indicating that intrinsic vessel aging is superimposed on metabolic effects to promote vascular disease. HGPS and WS serve as important examples to understand any causal relationship between vascular disease and aging.

    Because atherosclerosis compounds the pathological changes associated with normal vascular aging and vascular aging promotes atherosclerosis, this brings up the “chicken and egg” argument—does biological aging promote atherosclerosis, or does atherosclerosis promote vessel aging and cellular senescence? We suggest that these scenarios are not mutually exclusive but both occur simultaneously. While both structural changes associated with aging and cellular senescence are associated with atherosclerosis, this review focuses predominately on the latter, particularly on VSMC senescence, although other cell types undergo the same processes after the same stimuli. We will outline evidence of cellular senescence, proposed mechanisms, as well as current therapeutic options targeting vascular aging or cellular senescence in atherosclerosis.

    Evidence of Cellular Senescence in Atherosclerosis

    There are marked changes in multiple cell types in older versus younger individuals, including VSMCs,14,15 ECs,16 and inflammatory cells.17 Prematurely and naturally aged cells share several characteristics, including changes in cell proliferative potential, markers of cell senescence, increased propensity to undergo cell death, elevated DNA damage, and extensive telomere shortening and dysfunction. All of these features can be detected in cells from atherosclerotic plaques, which show additional features of cell senescence.

    Cell Proliferation

    VSMCs in the normal vessel wall have a very low turnover, with barely measurable proliferation indices. Increased cell proliferation is observed during early atherogenesis and on vascular injury,18 and aged VSMCs from rodents also show increased proliferation14,15,19 compared with cells from younger animals. In contrast, human VSMCs derived from both aged vessels and advanced atherosclerotic plaques undergo reduced proliferation and prolonged population doubling times.20,21 This observation corresponds to in vitro findings in which plaque VSMCs in culture show decreased percentages in S-phase and increased percentages in G1, consistent with a G1 growth arrest.21 While some of the arrest is associated with reduction in responses to mitogens, such as insulin-like growth factor 1 (IGF-1),22 the arrest is mediated by major changes in the expression of various cell cycle regulators, especially those involved in G1-S transition. Thus, increased expression of the cyclin-dependent kinase inhibitors p16INK4a and p21,23 decreased cyclin D and cyclin E,24 and hypophosphorylation of the retinoblastoma protein (pRB)2325 are observed in both normal human VSMCs undergoing replicative senescence and human plaque VSMCs. Plaque VSMCs also show reduced expression of the transcription factors E2F1–3 and increased sequestration of E2F1 to pRB.23 Importantly, these cell cycle regulators become potential markers of vascular cell senescence.

    Although most of these studies have examined cells that are presumed to be derived from the vessel wall itself, a number of published works suggest that at least a proportion of cells expressing VSMC or EC markers may be derived from bone marrow–derived progenitor cells (BMCs) and endothelial progenitor cells (EPCs) that migrate and integrate into the vessel wall and may themselves be affected by aging.26,27 This is a very controversial area,2830 but dysfunctional EPCs31 and impaired BMC migration and adhesion27,31 are seen in both aged and atherosclerotic mouse models. These cells from aged animals are accompanied by reduced expression of cell surface markers and cytokines for chemotaxis, such as C-X-C chemokine receptor type 4,27,32 decreased hypoxia-inducible factor 1A,33,34 as well as increased oxidative stress and inflammation.35,36

    Cell Senescence

    Cell senescence is defined as the irreversible loss of the ability of cells to divide. There are 2 general types of cell senescence, replicative senescence and stress-induced premature senescence (SIPS). Replicative senescence occurs with exhaustion of proliferative lifespan over time, a characteristic of aging, and is associated with critically shortened telomeres at chromosomal ends, which then induce a DNA damage response (DDR) (see below). In contrast, SIPS is triggered by external stimuli, including oxidizing agents and radiation, which activate the intracellular senescence cascade prematurely. While SIPS shares many morphological and molecular characteristics to replicative senescence, SIPS is not usually characterized by telomere shortening.

    As well as altered expression of cell cycle regulators, senescent cells are characterized by “specific” markers, including senescence-associated β galactosidase (SAβG), a lysosomal enzyme seen in senescence of multiple human cell types.37 Increased numbers of SAβG-positive VSMCs, ECs, and monocyte/macrophages are observed in aged vessels and atherosclerotic lesions when compared with their respective young and normal counterparts,23,38 reinforcing the idea that atherosclerosis is associated with premature cellular senescence. However, a word of caution is required when interpreting SAβG staining. In particular, cells with a high lysosomal content, such as macrophage foam cells, show SAβG reactivity that may not reflect senescence.

    Apoptosis

    Although cell death through multiple processes (necrosis, autophagy, apoptosis) occurs in atherosclerosis, the best described in association with senescence is apoptosis. Apoptosis, identified by morphological and molecular changes, is increased in aged vascular cells and is also increased in VSMCs and other cells in atherosclerotic plaques.18,21 While apoptosis occurs in ECs, VSMCs, and macrophages, most cell death in plaques is within the macrophage-rich necrotic core of the plaque. However, many apoptotic cells lose their lineage markers, which makes their precise identity difficult to be determined.18

    Telomere Shortening

    Telomeres consist of tandem repeats of the sequence TTAGGG at the end of chromosomes. At birth, telomere lengths in most tissues from the same subject are similar.39 Due to the mechanism of DNA replication, telomeres shorten with each cell division in cells that lack high activity of specific enzymes, such as telomerase, which maintain telomere length. Accelerated telomere shortening is also associated with dysregulation and dysfunction of proteins involved in telomere maintenance.40 Telomere length thus can be used to track a tissue's replication history and has been used as a surrogate for “biological” age.

    Telomere length and integrity is regulated through the interplay between telomerase, an enzyme composed of an RNA template (TERC) and catalytic subunit (TERT), and a complex of proteins known as Shelterin, consisting of TRF1, TRF2, Rap1, TIN2, PTOP1, and POT1,41 which pack the telomere into a so-called “T-loop.” The compact T-loop structure blocks the access of endonucleases required for telomere recombination42 and of DDR proteins that read telomeres as DNA double-stranded breaks.43 The T-loop can also regulate the access of telomerase to telomeres without affecting its expression and activity.4446 Indeed, increasing evidence suggests that telomere integrity rather than telomere length is responsible for controlling cell longevity.47 In particular, TRF2, a member of the Shelterin complex, may be a master regulator stabilizing the complex at telomeres and maintaining the compact T-loop. Cells overexpressing TRF2 continue to proliferate regardless of telomere length in vitro.47,48 In contrast, cells with mutated TRF2 show aberrant chromosome structures, including chromosome end-fusions and shorter telomeres, which are sufficient to induce premature senescence and apoptosis.4850

    Shortened telomeres are evident in atherosclerosis, observed in plaque VSMCs23 and ECs51 relative to the normal vessel wall, and in circulating EPCs.36 Also, telomeres in leukocytes are shorter in patients with atherosclerosis compared with control subjects52,53 and are inversely correlated to cardiovascular disease risks in patients with subclinical diseases.54,55 Short telomeres and low levels of telomerase expression and activity are functionally important in VSMC senescence, as ectopic telomerase expression can dramatically increase lifespan of both plaque and normal VSMCs.23 However, some of these effects may be independent of telomeres, as telomeres continue to shorten in these cells and cells replicate with critically short telomeres.23 In addition, whereas telomere length mostly reflects previous replication, arterial segments resistant to atherosclerosis, such as internal mammary artery or ascending aorta, have longer telomeres than the aortic regions prone to the disease.56,57 This difference is age-independent, suggesting the existence of intrinsic genetic or developmental variations in telomere regulation may underlie location-specific predisposition in atherogenesis.

    The ease that telomere length can be measured has resulted in an explosion in studies associating telomere length with multiple diseases or preclinical disease. However, there are important caveats to the slavish adoption of telomere length as a biomarker for aging in disease, or in animal models. Importantly, telomere length is relatively stable over time between 20 to 70 years of age in humans when vascular aging is apparent; telomere length rarely reaches levels classified as “critical” to affect telomere function in cardiovascular studies, making it unclear whether the cells with shorter telomere length have any discernable loss of function; and telomeres in animal models (especially rodents) have a very different structure to those in humans. Finally, unlike other biomarkers of disease activity, successful treatment of cardiovascular disease is highly unlikely to increase telomere length, although it might slow loss of telomeres.

    DNA Damage

    Aside from shortened telomeres, VSMCs, ECs, macrophages, and circulating cells from the elderly and patients with atherosclerosis contain increased DNA damage in both nuclei and mitochondria5861 compared with younger subjects and those without vascular disease. The accumulation of DNA damage in cells may reflect both ongoing damage-inducing stimuli and defects in the repair machinery. Similar to telomere shortening, DNA damage eventually leads to cell senescence and apoptosis, although minor damage is associated with transient growth arrest as repair is undertaken. DNA damage includes both single- and double-stranded breaks, deleted sections of DNA, nucleotide modifications, and extrusions of DNA from the nucleus (micronuclei), accompanied by altered expression of various DDR proteins.62 In brief, the DDR is initiated by the identification of “faulty” DNA, either a mismatch base or single- or double-stranded breaks. Identification and signaling is achieved by recruiting various sensor proteins, including phosphorylated forms of ataxia telangiectasia mutated (P-ATM) and histone 2A protein X (γ-H2AX) (Figure 2). Expression of both P-ATM and γ-H2AX increase with atherosclerotic plaque grade in vivo, and both markers are elevated in plaque VSMCs in vitro compared with cells from normal vessels.61 P-ATM and γ-H2AX are therefore used as common markers to identify DNA damage and the DDR in vivo. Recruitment of these proteins triggers an intracellular kinase cascade leading to the activation of a range of effectors to participate in DNA repair, including p53 and Chk2. The normal response is transient cell cycle arrest and repair of the damaged DNA. However, when DNA damage is too extensive to be repaired or when the repairing cascades are impaired, cell senescence and apoptosis occur.

    Figure 2.

    Figure 2. Model of premature cellular senescence in atherosclerosis. Cardiovascular risk factors including genetic predisposition accelerate normal biological aging of vessels, resulting in telomere attrition, accumulated DNA damage, increased oxidative stress, and epigenetic modifications. Inherited defects in DNA repair enzymes or lamin also accelerate this process. While epigenetic modifications lead to transcriptional alterations that can change cell function, activation of the DNA damage response (DDR) is a common result. DDR sensors, such as ATM and H2AX, are phosphorylated and bind to damaged DNA regions, followed by recruitment of various DDR proteins including MRE11 and NBS1. These signaling pathways activate various downstream effectors, such as p53 and Chk2, leading to temporary growth arrest for DNA damage repair. Successful repair allows cells to continue proliferating and repair vessel damage. In contrast, unsuccessful DNA repair results in accumulation of both nuclear and mitochondrial DNA damage, and cells undergo telomere-dependent and telomere-independent senescence and apoptosis and secrete proinflammatory cytokines as part of the SASP. These processes lead to the loss of normal cellular function and increased inflammation seen in aging and in atherosclerotic plaques. Cellular dysfunction contributes to the alterations in ECM proteins, resulting in vascular stiffness and loss of elasticity, and the proinflammatory state promotes atherosclerosis. Atherosclerosis may also contribute directly to accelerated vascular aging, as vascular repair can promote replicative senescence, and the proinflammatory state and ROS promote stress-induced premature senescence (SIPS) (illustration: Cosmocyte/Ben Smith).

    DNA damage in mitochondria is also seen in atherosclerosis, characterized by a 4977 bp deletion.60 This deletion is associated with mitochondrial dysfunction,63 although it is not clear whether the levels of the deletion observed in atherosclerosis are sufficient alone to cause mitochondrial dysfunction. Whereas gradual loss of mitochondrial function occurs with age, it can be accelerated by oxidative stress, for example due to increased oxidized LDL (oxLDL) during atherogenesis.64,65

    Epigenetic Modifications

    Epigenetic modifications, including histone acetylation and DNA methylation, refer to a postreplication alteration of chromatin without affecting the basal nucleotide code. Epigenetic mechanisms are important in regulating gene silencing and activation. Age-related changes in methylation and acetylation patterns in the genome have been reviewed intensively66,67 and are linked to various diseases of premature aging, including atherosclerosis.68 Epigenetic changes are therefore used increasingly as markers of premature cellular senescence in the vasculature.

    Although DNA methylation patterns are heterogeneous between sex, race, and age69,70 that make them challenging to use as biomarkers for atherogenesis, recent studies have identified changes in the expression of methyltransferases and hence in methylation in plaques and leukocytes from both patients and atherogenic mice.7173 Altered methyltransferase expression correlates with hypomethylation of normally hypermethylated genomic regions and vice versa, which can occur within genes participating in proliferation, apoptosis and lipid metabolism. For example, hypomethylation corresponds with phenotypic modulation and proliferation in VSMCs71; changes in methylation occur before overt atherosclerosis in mice models; and atherogenic lipoproteins can enhance DNA methylation and histone deacetylation.74 These findings suggest a potential feedback loop between epigenetic modification and atherosclerosis.

    Histone acetylation and deacetylation are also important contributors to atherosclerosis and aging, through which inflammation, VSMC proliferation, and ECM composition can be modulated. A well-studied example is the mitochondrial gene p66(Shc), also known as a longevity gene. Increased hypermethylation and histone acetylation result in the age-related enhancement of p66Shc production and hence the subsequent availability for activation.75 In contrast, deletion of p66(Shc) is atheroprotective by reducing oxidative stress, endothelial dysfunction, and vascular cell apoptosis.76,77

    Mechanisms of Cellular Senescence in Atherosclerosis

    The mechanisms underlying cellular senescence in atherosclerosis are likely to be multiple and cumulative in any one cell or artery. For example, telomere shortening leading to replicative senescence will interact with nuclear and mitochondrial DNA damage due to free radicals, which themselves will be increased where there are defects in DNA repair. DNA damage may also interact with epigenetic modifications to regulate genes that themselves regulate cell proliferation, cell senescence, and apoptosis. For simplicity, we will outline each mechanism in turn and also provide evidence that each may be causal in atherosclerosis.

    Telomere Maintenance

    As described above, telomere shortening in vascular cells occurs with replication and is seen in plaques. Telomere shortening of circulating leukocytes also occurs with aging and in atherosclerosis, probably due to gradual bone marrow failure with telomere shortening of progenitor cells.78 Telomere length may therefore have functional consequences in aging and atherosclerosis. In contrast, whereas ectopic telomerase can maintain cell proliferation23 and reverse the tissue degeneration in TERT-deficient mice,79 reduction of telomerase activity in TERC−/− mice was found to be atheroprotective,80 linked to decreased proliferative potential in macrophages and other inflammatory cells. These opposing findings suggest that telomerase activity may affect atherosclerotic development in a stage-dependent manner or that manipulation of telomere length by associated proteins may affect different cells differently. Indeed, dysfunction of the Shelterin protein complex has different effects on macrophages compared with lymphocytes.81 In addition, telomerase overexpression cannot always prevent and/or delay cell senescence, as observed in both VSMCs and ECs.82,83 This suggests that telomere-independent mechanisms, such as increase in genomic instability and mitochondria dysfunction, may contribute to the effects of DNA damage in atherosclerosis.

    DNA Damage Recognition and Repair

    DNA damage occurs naturally by both intrinsic (for example, spontaneous mutation and replication errors) and extrinsic (for instance, UV radiation, chemical exposure, and free radicals) stimuli. Although DNA damage and DDR activation are seen in atherosclerosis, increasing evidence from studies of DDR proteins and repair enzymes indicates that DNA damage directly promotes both atherosclerosis and medial degeneration. For example, ATM heterozygosity accelerates atherosclerosis in mouse models, accompanied by increased nuclear and mitochondrial DNA damage and metabolic changes similar to the metabolic syndrome.63,84 These phenotypes can be partially rescued by bone marrow transplant with wild-type ATM. Similarly, age-associated accumulation of prelamin A disrupts normal compartmentalization of nuclear signaling pathways and leads to insufficient DNA repair, which in part contributes to DNA damage accumulation and accelerates atherosclerosis.85 As described above, defects in lamin A are also found in HGPS patients, whose cells contain increased DNA damage and who show signs of premature aging including atherosclerosis.86,87 Moreover, cells from patients with WS and the representative knock-out animal models show genetic instability.88,89 Thus, defective DNA repair directly promotes atherosclerosis.

    Reactive Oxygen Species and Oxidative Stress

    In vitro studies have shown that in comparison to VSMCs from younger subjects, aged VSMCs contained greater oxidative stress-induced damage, probably due to the combination of higher reactive oxygen species (ROS) generation and impaired antioxidant defense.61,90,91 The major source of ROS within the cell is as a by-product of oxidative phosphorylation. Mitochondrial DNA damage and dysfunction can increase ROS generation, creating a positive feedback loop. ROS are also increased by elevated levels of oxidized lipoproteins in atherosclerosis, with the most common form being reactive hydroxyl free radicals (OH). Moreover, ROS production can be regulated through mechanical stress, as ECs under laminar shear stress upregulate antioxidant enzymes such as peroxiredoxins.92 Turbulent flow at sites of plaque formation would thus reduce antioxidant capacity.

    ROS promote DNA damage by either adding double bonds to or removing the hydrogen atom from DNA bases. Oxidative DNA damage occurs in both mitochondrial and nDNA and in both telomeric and nontelomeric regions.59,65 Indeed, the level of 8-oxo-deoxyguanosine (8-oxodG), an oxidized form of guanine marking oxidative DNA damage, is elevated in VSMCs and macrophages in plaques,23,59 and, as discussed earlier, increased DNA damage and the subsequent DDR promote cell senescence and apoptosis. In addition, at higher concentrations, ROS can produce SIPS in vascular cells.23,61

    Apart from directly promoting DNA damage, ROS can modulate atherosclerosis progression by other mechanisms that also involve premature cellular senescence. For example, ROS can inactivate the PI-3K/Akt pathway,93 which can downregulate and inactivate telomerase94 and directly promote apoptosis.95 ROS-induced p53 activation, possibly indirectly via increasing the DDR, reduces the expression of the IGF-1 receptor in VSMCs and the subsequent Akt survival signals.95 ROS can also stimulate transforming growth factor-β (TGFβ) signaling by inducing the release of active TGFβ from its inactive secreted form, the latent transforming growth factor-β (LTGFβ).96 Furthermore, ROS can increase the expression of cell surface receptors required for uptake of oxLDL,97 a feature also seen in aging.

    Epigenetic Modifications

    Increasing evidence suggests that not only do epigenetic modulations involving changes in methylation, acetylation, and noncoding RNA occur in atherosclerosis, these changes regulate important processes in premature cellular senescence and atherosclerosis. For example, the monocarboxylate transporter (MCT)3, which participates in lactate transport in VSMCs, is hypermethylated in atherosclerotic lesions.98 As a consequence, MCT3 expression is reduced and is associated with VSMC proliferation. Similarly, estrogen receptors (ER) are expressed in both ECs and VSMCs.99 Hypermethylation of both receptor subunits is detected with normal aging and in patients with atherosclerosis100,101 and correlates with ER abundance.99 In particular, hypermethylation of ERα leads to reduced expression that might contribute to VSMC proliferation in early atherosclerosis,100,102 whereas hypermethylation of ERβ appears in advanced lesions and is involved in vascular senescence.101

    A number of miRNAs are upregulated in atherosclerosis and/or regulate processes associated with aging, such as cell proliferation, apoptosis, or senescence (summarized in Figure 3). For example, miR-21 is increased after vessel injury, promotes VSMC proliferation and regulates VSMC survival through modulation of phosphatase and tensin homolog and B-cell lymphoma-2 (Bcl-2) expression.103 Similarly, levels of miR-221/222 are elevated both in patients with coronary artery disease104 and after vessel injury.105 miR-221/222 is upregulated by PDGF and also promotes VSMC proliferation, in part by downregulation of p27kip.105,106 In contrast, the expression of miR-143/145 which are controlled by TGFβ signaling107 are repressed during neointimal formation on injury and in atherosclerotic development.108 Increased TGFβ in advanced stages of atherosclerosis enhances miR-143/145 expression, which subsequently inhibits Kruppel-like factor-4/5 (KLF4/5) and hence prohibits VSMC proliferation.107,109 Increased cell cycle arrest and senescence in VSMCs can promote miR-133, which further represses VSMC proliferative phenotype and forms a feedback loop. In addition, overexpressing miR-143/145 at early point of vascular injury can limit neointimal formation,108 further strengthening their importance in vascular remodeling. Micro (Mi)RNAs are also extensively involved in regulating ECs.

    Figure 3.

    Figure 3. miRNAs regulate cell proliferation, cell senescence, and apoptosis through multiple pathways. Mitogens such as PDGF induce activation of miR-21, miR-221, and miR-222, leading to cell survival (through Bcl2 and phosphatase and tensin homolog [PTEN]) and cell proliferation via inhibition of p27kip1. Ultimately, this may promote VSMC proliferation, for example, in early atherosclerosis or in vascular repair. In contrast, VSMCs from aged vessels in atherosclerotic plaques show reduced responses to mitogens accompanied by activation of TGFβ signaling pathways, leading to the upregulation of multiple miRNAs that inhibit VSMC proliferation and promote cellular senescence, features that would be predicted to promote plaque rupture and/or inhibit plaque repair.

    For example, senescent ECs show reduced expression of miR-21, miR-214, and miR-92, accompanied by increased miR-221/222.110 Moreover, miRNAs can also modulate atherosclerosis by targeting genes involved in lipid metabolism. For instance, miR-33 suppresses the expression of the cholesterol transporter ABCA1, leading to reduced high-density lipoprotein (HDL) biogenesis111 and fatty acid oxidation.112 Antagonizing miR-33 activity is atheroprotective, accompanied by increased HDL and smaller plaques.111 Although these examples of changes in epigenetic markers and miRNA abundance are interesting, what is lacking is direct evidence that either is causally responsible for the changes in cell proliferation or senescence in normal or premature aging. This is clearly a future area for study.

    Systemic Causes

    The above discussion has focused on local stimuli for cellular senescence and vascular aging, outlining the cellular mechanisms involved. However, there are many systemic factors that predispose to vascular aging, including genetic predisposition, some of which are also risk factors for atherosclerosis. It is now becoming clear that their effects are mediated at least in part through accelerated cellular senescence and through the mechanisms outlined.

    Arterial aging has a significant genetic predisposition and varies between races. For example, African-Caribbean individuals have accelerated arterial aging, increased aortic stiffness and greater carotid intima thickness when compared with European and Asian populations.113115 This indicates the likely presence of genetic loci controlling multiple aspects of vessel aging. Similarly, age-related artery stiffness is more pronounced in women and is independent of the menopause, reflecting intrinsic differences in arterial properties between sexes.116,117 Although men have more prominent accelerated aging over time compared with women,7 there are complex effects of hormone deficiency and replacement therapy on arterial stiffness and incidence of nonfatal cardiovascular diseases118,119 that may be reversed on withdrawal.120 Thus, both genetic predisposition and sex affect vessel aging.

    Acquired risk factors are also major contributors to premature vascular aging and cellular senescence. For example, smoking increases arterial stiffness and pulse pressure121 and promotes SIPS in ECs and fibroblasts.1,122 Similarly, whereas low to moderate and/or acute alcohol intake are associated with reduced arterial stiffness, chronic higher alcohol intake (>21 U/wk) results in opposing effects.123,124 Finally, age- and diet-associated elevation of serum glucose is associated with both premature vascular stiffness and vascular cell senescence, which are particularly pronounced in younger patients125 and can be reduced by physiological levels of insulin.126 Glucose participates in a spontaneous, nonenzymatic reaction known as glycation, which adds sugars to proteins and lipids to form reactive advanced glycation end products (AGEs).6 Both glycation and AGEs increase with age and in patients with atherosclerosis and metabolic disorders and correlate with serum glucose levels.127 Multiple animal studies demonstrate the underlying mechanisms through which vascular aging is promoted by glycation,128 including promoting vascular stiffness through collagen stabilization129 and attenuating nitric oxide (NO) production in ECs,130 increasing vascular permeability and cell adhesion,131,132 enhancing VSMC migration,133 augmenting circulating lipoproteins for oxidation and their subsequent uptake by macrophages,134 and increasing inflammation and oxidative stress.131,135,136 Reducing dietary AGEs can be atheroprotective137,138 and glycation can be modulated by other risks factors, for example by smoking.139

    Functional Consequences of Cell Aging

    Aging-associated changes are observed in multiple cell types and are conserved across species, from rodents to primates. As well as loss of normal cellular function, these changes result in specific consequences in each cell type, which may be directly proatherogenic.

    Endothelial Cells

    Aged ECs become flatter, enlarged, and have an increasingly polypoid nucleus, all features associated with cellular senescence.140143 These changes are accompanied by modulation in cytoskeleton integrity, proliferation, angiogenesis, and cell migration. Senescent ECs show attenuated endothelial NO production144 and increased endothelin-1 release.145 Late-passage ECs also show reduced expression of the adhesion molecules VCAM-1 and intracellular adhesion molecule-1 (ICAM-1), increased activation of nuclear factor (NF)-κB, and increased susceptibility to apoptosis.143 Moreover, there are marked age-associated changes in ICAM-1 function and activity.146 Thus, EC senescence is associated with loss of EC function and a shift toward a proinflammatory and proapoptotic state, predicted to enhance monocyte migration into the vessel wall.

    Vascular Smooth Muscle Cells

    VSMCs in human plaques or derived from plaques show reduced proliferation, early senescence, and increased susceptibility to apoptosis. These properties would reduce the ability to repair plaques that undergo rupture. Aged rodent aortas also show increased levels of interleukin (IL)-6 and aged aortic VSMCs have a higher basal secretion of IL-6 than young VSMCs. Indeed, secretion of a common set of secreted proteins as cells age is a widespread phenomenon known as the “senescence-associated secretory phenotype,” or SASP. Moreover, aged VSMCs exhibit upregulation of chemokines (chemokine CC-motif ligand 2), adhesion molecules (eg, ICAM-1), and innate immune receptors (eg, Toll-like receptor 4).147 These properties generate a proinflammatory environment, further promoting migration of inflammatory cells. VSMC senescence thus promotes atherosclerosis progression and inhibits plaque repair.

    Inflammatory Cells

    Monocytes from patients with atherosclerosis demonstrate an increased burst of free radicals on activation and increased secretion of a number of cytokines, including monocyte chemotactic protein (MCP)-1, IL-6, IL-1β, and tumor necrosis factor-α.81 Importantly, many of these differences are also observed in aged versus young monocytes and can be recapitulated by agents that disrupt telomeres.81 Thus, there is direct evidence that aging promotes proinflammatory changes in monocyte/macrophages that are relevant to atherosclerosis.81 Coupled with altered adhesion molecules on aged ECs, aging would be predicted to promote both migration and activation of macrophages within plaques.

    Targeting Aging in Atherosclerosis

    The association between aging and atherosclerosis and the increasing evidence demonstrating that accelerated cellular senescence occurs in vascular disease have resulted in the search for treatments that can promote longevity and delay senescence. Indeed, pharmaceutical companies are now developing drugs that target specific mechanisms of premature aging, including telomerase activity, DNA methylation, DNA damage repair, and miRNA. Specific agents may also target the DNA damage machinery and DDR directly. For example, chloroquine has been demonstrated in animal models to reduce atherosclerosis through a p53-dependent ATM signaling pathway.148 Similarly, pioglitazone, a peroxisome proliferator-activated receptor agonist, can increase telomerase activity, TRF-2 expression, and phosphorylation of Akt and reduce the expression of the senescence markers p16, cell-cycle checkpoint kinase 2, and p53.149

    Currently, it is debatable whether telomere length and telomerase are targets in atherosclerosis. As described above, although atherosclerosis is associated with telomere shortening in multiple cell types, apart from VSMCs23 there is a paucity of evidence to demonstrate that shortening occurs to critical lengths that impair function. There is also only a little evidence81 to show that the telomere shortening in vivo makes leukocytes proatherosclerotic. Furthermore, there is minimal evidence to suggest that telomere shortening initiates atherosclerosis rather than being a feature of advanced plaques. Although the lifespan of VSMCs and ECs can be extended by ectopic expression of telomerase, it is not clear if this is due to effects only on telomere length, and the fact that plaque VSMCs cannot be extended indefinitely indicates that plaque VSMCs have other causes for their senescence.23 In addition, as described above, the mouse studies of telomerase manipulation in atherosclerosis are contradictory. Finally, and most importantly, increasing telomerase expression systemically has considerable risks. Agents that inhibit telomerase are being developed for cancer,150 indicating that augmenting telomerase has the potential to be carcinogenic and can add the undesirability of uncontrolled proliferation of VSMCs. Therefore, much more research must be undertaken in this area before manipulation of telomere length or telomerase activity as a primary mode of action could be considered for therapeutics in atherosclerosis, although agents that indirectly promote telomere stability in cells on the vessel wall may still be beneficial.

    In addition to more specific mechanisms, a number of currently available drugs and compounds are likely to delay premature aging as part of their mechanisms of action through changes in ROS and oxidative DNA damage. Examples of these include antioxidants, statins, and angiotensin-converting enzyme (ACE) inhibitors/angiotensin receptor blockers (ARBs). Dietary manipulation and augmentation of sirtuin activity also have potential to reduce vascular aging, cellular senescence, and atherosclerosis.

    Antioxidants

    Expression of naturally occurring antioxidants including ferritin and glutathione are reduced with age and in atherosclerosis.151,152 Coupled with evidence of increased ROS in atherosclerosis and the demonstration that ROS promote premature aging, this has resulted in intensive study of the use of antioxidants to prevent cardiovascular diseases and aging. However, although antioxidants have been demonstrated to have significant antiatherosclerosis effects in vitro and in animal models, their efficacy in humans is questionable. For example, Vitamin E can reduce the uptake of oxLDL by macrophages and inhibit the subsequent inflammatory responses that stimulate atherosclerosis development.97,153 Also, antioxidants can modulate epigenetic patterns through regulating levels of methyl donors and methyltransferase inhibitors.154 However, there is now extensive evidence indicating that supplementing dietary antioxidants has no significant effect on reducing cardiovascular risk.155,156

    Although there may be many reasons why antioxidants might not reduce atherosclerosis, the therapeutic failure of dietary supplementation has simulated interest in enhancing natural antioxidant pathways. For example, overexpression of nuclear factor E2–related factor-2 (Nrf2), a key transcription factor orchestrating antioxidant and cytoprotective responses, in VSMCs and ECs reduces oxidative stress and attenuates inflammatory responses.157,158 Nrf2 activators such as Protandim can increase the production of multiple antioxidant enzymes.159 In contrast, Nrf2 knockout mice develop smaller atherosclerotic plaques with improved arterial stiffness, regardless of increased serum cholesterol and lipid oxidation.160 This phenotype is achieved, at least in part, through downregulation of the CD36 receptor required for oxLDL uptake by macrophages, which reduces macrophage apoptosis and the subsequent inflammatory responses. These findings demonstrate that Nrf2 mediates multiple pathways independent of its antioxidant effects, and more studies are required before using Nrf2 as a therapeutic option for atherosclerosis.

    Statins

    The hydroxy-methylglutaryl-coenzyme A reductase inhibitors (statins) reduce cholesterol synthesis and hence reduce atherogenesis, the progression of established plaques, and cardiovascular risk. Although their major effect may be via cholesterol reduction, statins have multiple effects that are not necessarily associated with their primary mechanism, including improved endothelial function and reduced inflammation responses. More specifically, statins target mechanisms inducing premature aging, leading to enhanced telomere protection through upregulating TRF2,161 decreased DNA damage by accelerating DNA damage repair,61,162 and suppressing oxidative stress in part by increasing antioxidant defenses.163 Statins can also delay VSMC replicative senescence and reduce markers of DNA damage in vivo in atherosclerosis.61

    ACE Inhibitors/ARBs

    Angiotensin II increases ROS and promotes oxidative DNA damage, promoting senescence through telomeric and nontelomeric DNA damage.82 Whereas ACE inhibitors/ARBs have multiple cardiovascular effects, they can also reduce oxidative stress and subsequent DNA damage. At present, it is not known whether these drugs reduce premature aging in vivo and whether this is an important mode of their action. However, bradykinin, a hormone that mediates some of the vasoprotective effects of ACE inhibitors, protects ECs from superoxide-induced senescence through bradykinin B2 receptor-mediated and NO-mediated inhibition of DNA damage.164

    Dietary Manipulation and Sirtuins

    Nutritional status and diet are associated with age-related vascular changes and with atherosclerosis.165 In particular, caloric restriction (CR) is a common method to manipulate diet and the beneficial effects of CR on vessel aging have been proven in animal models,166168 including preservation of matrix components within the vessel wall,169 improving EC function through augmenting NO generation,170 reducing sensitivity to oxLDL,171 reducing oxidative stress by upregulating antioxidants and protecting mitochondria function,170,172 and inhibiting inflammation.173 Although CR may be inappropriate in many patients, drugs and dietary supplements that mimic CR effects without affecting nutritional balance may offer a wider therapeutic option.

    Sirtuins (SIRT1–7) are a family of nicotinamide adenine dinucleotide (NAD)+-dependent deacetylases and adenosime diphosphate–ribosyltransferases that may be partially responsible for the age-delaying effects of CR. Sirtuins have been reviewed as part of this series174; therefore only limited discussion is provided here, focusing on SIRT1, the most well-studied member. CR increases SIRT1 in some experimental models, leading to improved endothelial function167,168 while knocking down SIRT1 interferes with the CR-mediated antioxidant and anti-inflammatory vascular effects.173 Similar to CR, overexpression of SIRT1 in the endothelium can improve vascular stiffness and attenuate the development of atherosclerosis,175 probably by activating endothelial nitric oxide synthase (eNOS) and promoting NO production175,176 and preventing EC senescence.177 Indeed, SIRT deacetylation of eNOS may contribute to the atheroprotective effects of laminar stress.178 SIRT1 in hematopoietic cells also prevents foam cell formation and reduces atherosclerosis.179 The agent Resveratrol, which can increase SIRT1 expression, reduces EC apoptosis and increases aortic elasticity in aged rodents,180 although how much of this effect is due to SIRT1 is unclear. Moreover, other sirtuin members, such as SIRT3–5, as sensors of nutritional status may be protective in vascular system, regulating the cellular response to stress, energy production, apoptosis, and ROS production.181,182

    Conclusions

    In summary, both normal vascular aging and atherosclerosis are associated with cellular senescence. Cellular senescence impairs cell proliferation resulting in irreversible growth arrest and impairs survival, due to an accumulation of nuclear and mitochondrial DNA damage, increased ROS, and a proinflammatory state. Both vascular aging and cellular senescence are associated with increased expression of proinflammatory cytokines and adhesion molecules further promoting inflammation and also affect the synthesis and maintenance of extracellular matrix proteins. Aging can be identified by both structural changes and by a number of senescence-associated biomarkers. However, major gaps in our knowledge exist as to whether small changes in these biomarkers reflect an important loss of function and how aged cells promote diseases. As advanced atherosclerosis is likely to manifest irreversible changes, prevention of accelerated cell aging becomes a major therapeutic opportunity. Understanding the mechanisms contributing to such changes is therefore crucial for both the prevention and the development of treatment for atherosclerosis and other age-related diseases.

    Non-standard Abbreviations and Acronyms

    ACE

    angiotensin-converting enzyme

    AGE

    advanced glycation end products

    ARB

    angiotensin receptor blocker

    ATM

    ataxia telangiectasia mutated

    Bcl-2

    B-cell lymphoma-2

    BMC

    bone marrow–derived progenitor cell

    CR

    caloric restriction

    DDR

    DNA damage response

    EC

    endothelial cell

    ECM

    extracellular matrix

    eNOS

    endothelial nitric oxide synthase

    EPC

    endothelial progenitor cell

    ER

    estrogen receptor

    H2AX

    histone 2A protein X

    HDL

    high-density lipoprotein

    HGPS

    Hutchinson-Gilford progeria syndrome

    ICAM-1

    intracellular adhesion molecule-1

    IGF-1

    insulin-like growth factor-1

    IL

    interleukin

    LDL

    low-density lipoprotein

    LTGFβ

    latent transforming growth factor-β

    KLF

    Kruppel-like factor

    MCP

    monocyte chemotactic protein

    MCT3

    monocarboxylate transporter 3

    MiRNA/miR

    Micro RNA

    MRE11

    meiotic recombination 11

    NAD+

    nicotinamide adenine dinucleotide

    NBS1

    Nijmegen breakage syndrome-1

    NF-κB

    nuclear factor-κB

    NO

    nitric oxide

    Nrf2

    nuclear factor E2-related factor-2

    OxLDL

    oxidized low-density lipoprotein

    PDGF

    platelet-derived growth factor

    ROS

    reactive oxygen species

    POT1

    protection of telomeres protein 1

    pRB

    retinoblastoma protein

    SAβG

    senescence-associated β-galactosidase

    SASP

    senescence-associated secretory phenotype

    SIPS

    stress-induced premature senescence

    TERC

    telomerase RNA component

    TERT

    telomerase reverse transcriptase

    TGFβ

    transforming growth factor-β

    TIN2

    TRF1-interacting nuclear factor 2

    TLR4

    Toll-like receptor 4

    TPP1

    tripeptidyl teptidase I

    TRF1

    telomeric repeat binding factor-1

    TRF2

    telomeric repeat binding factor-2

    VCAM-1

    vascular cell adhesion molecule-1

    VSMC

    vascular smooth muscle cell

    WRN

    Werner syndrome RecQ helicase

    WS

    Werner syndrome

    Sources of Funding

    This work was supported by British Heart Foundation Grant RG/08/009/25841 and the NIHR Cambridge Biomedical Research Centre .

    Disclosures

    None.

    Footnotes

    In May 2012, the average time from submission to first decision for all original research papers submitted to Circulation Research was 12.0 days.

    Correspondence to Martin Bennett, MD, PhD,
    Division of Cardiovascular Medicine, University of Cambridge, Box 110, ACCI, Addenbrooke's Hospital, Cambridge, CB2 0QQ, UK
    . E-mail

    References

    • 1. Farhat N, Thorin-Trescases N, Voghel G, Villeneuve L, Mamarbachi M, Perrault LP, Carrier M, Thorin E . Stress-induced senescence predominates in endothelial cells isolated from atherosclerotic chronic smokers. Can J Physiol Pharmacol. 2008; 86: 761–769.CrossrefMedlineGoogle Scholar
    • 2. Niemann B, Chen Y, Teschner M, Li L, Silber RE, Rohrbach S . Obesity induces signs of premature cardiac aging in younger patients: the role of mitochondria. J Am Coll Cardiol. 2011; 57: 577–585.CrossrefMedlineGoogle Scholar
    • 3. Bolton E, Rajkumar C . The ageing cardiovascular system. Rev Clinical Gerontol. 2010; 21: 99–109.CrossrefGoogle Scholar
    • 4. Virmani R, Avolio AP, Mergner WJ, Robinowitz M, Herderick EE, Cornhill JF, Guo SY, Liu TH, Ou DY, O'Rourke M . Effect of aging on aortic morphology in populations with high and low prevalence of hypertension and atherosclerosis: comparison between occidental and Chinese communities. Am J Pathol. 1991; 139: 1119–1129.MedlineGoogle Scholar
    • 5. O'Rourke MF, Hashimoto J . Mechanical factors in arterial aging: a clinical perspective. J Am Coll Cardiol. 2007; 50: 1–13.CrossrefMedlineGoogle Scholar
    • 6. Zieman S, Kass D . Advanced glycation end product cross-linking: pathophysiologic role and therapeutic target in cardiovascular disease. Congest Heart Fail. 2004; 10: 144–149.CrossrefMedlineGoogle Scholar
    • 7. Astrand H, Stalhand J, Karlsson J, Karlsson M, Sonesson B, Lanne T . In vivo estimation of the contribution of elastin and collagen to the mechanical properties in the human abdominal aorta: effect of age and sex. J Appl Physiol. 2011; 110: 176–187.CrossrefMedlineGoogle Scholar
    • 8. Franklin SS, Gustin Wt, Wong ND, Larson MG, Weber MA, Kannel WB, Levy D . Hemodynamic patterns of age-related changes in blood pressure: the Framingham Heart Study. Circulation. 1997; 96: 308–315.LinkGoogle Scholar
    • 9. Wang M, Zhang J, Jiang LQ, Spinetti G, Pintus G, Monticone R, Kolodgie FD, Virmani R, Lakatta EG . Proinflammatory profile within the grossly normal aged human aortic wall. Hypertension. 2007; 50: 219–227.LinkGoogle Scholar
    • 10. Hashimoto M, Ishinaga Y, Honda M, Ohoka M, Morioka S, Moriyama K . Age-related increase in the uptake of acetylated low density lipoprotein into cultured endothelial cells from rat aorta. Exp Gerontol. 1991; 26: 397–406.CrossrefMedlineGoogle Scholar
    • 11. Olive M, Harten I, Mitchell R , et al . Cardiovascular pathology in Hutchinson-Gilford progeria: correlation with the vascular pathology of aging. Arterioscler Thromb Vasc Biol. 2010; 30: 2301–2309.LinkGoogle Scholar
    • 12. Varga R, Eriksson M, Erdos MR , et al . Progressive vascular smooth muscle cell defects in a mouse model of Hutchinson-Gilford progeria syndrome. Proc Natl Acad Sci U S A. 2006; 103: 3250–3255.CrossrefMedlineGoogle Scholar
    • 13. Massip L, Garand C, Turaga RV, Deschenes F, Thorin E, Lebel M . Increased insulin, triglycerides, reactive oxygen species, and cardiac fibrosis in mice with a mutation in the helicase domain of the Werner syndrome gene homologue. Exp Gerontol. 2006; 41: 157–168.CrossrefMedlineGoogle Scholar
    • 14. Stemerman M, Weinstein R, Rowe J, Maciag T, Fuhro R, Gardner R . Vascular smooth muscle cell growth kinetics in vivo in aged rats. Proc Natl Acad Sci U S A. 1982; 79: 3863–3866.CrossrefMedlineGoogle Scholar
    • 15. Hariri RJ, Hajjar DP, Coletti D, Alonso DR, Weksler ME, Rabellino E . Aging and arteriosclerosis: cell cycle kinetics of young and old arterial smooth muscle cells. Am J Pathol. 1988; 131: 132–136.MedlineGoogle Scholar
    • 16. Gerhard M, Roddy MA, Creager SJ, Creager MA . Aging progressively impairs endothelium-dependent vasodilation in forearm resistance vessels of humans. Hypertension. 1996; 27: 849–853.LinkGoogle Scholar
    • 17. Chung HY, Sung B, Jung KJ, Zou Y, Yu BP . The molecular inflammatory process in aging. Antioxid Redox Signal. 2006; 8: 572–581.CrossrefMedlineGoogle Scholar
    • 18. Lutgens E, de Muinck ED, Kitslaar PJ, Tordoir JH, Wellens HJ, Daemen MJ . Biphasic pattern of cell turnover characterizes the progression from fatty streaks to ruptured human atherosclerotic plaques. Cardiovasc Res. 1999; 41: 473–479.CrossrefMedlineGoogle Scholar
    • 19. Moon SK, Cha BY, Kim CH . In vitro cellular aging is associated with enhanced proliferative capacity, G1 cell cycle modulation, and matrix metalloproteinase-9 regulation in mouse aortic smooth muscle cells. Arch Biochem Biophys. 2003; 418: 39–48.CrossrefMedlineGoogle Scholar
    • 20. O'Brien ER, Alpers CE, Stewart DK, Ferguson M, Tran N, Gordon D, Benditt EP, Hinohara T, Simpson JB, Schwartz SM . Proliferation in primary and restenotic coronary atherectomy tissue: implications for antiproliferative therapy. Circ Res. 1993; 73: 223–231.LinkGoogle Scholar
    • 21. Bennett MR, Evan GI, Schwartz SM . Apoptosis of human vascular smooth muscle cells derived from normal vessels and coronary atherosclerotic plaques. J Clin Invest. 1995; 95: 2266–2274.CrossrefMedlineGoogle Scholar
    • 22. Patel VA, Zhang QJ, Siddle K, Soos MA, Goddard M, Weissberg PL, Bennett MR . Defect in insulin-like growth factor-1 survival mechanism in atherosclerotic plaque-derived vascular smooth muscle cells is mediated by reduced surface binding and signaling. Circ Res. 2001; 88: 895–902.LinkGoogle Scholar
    • 23. Matthews C, Gorenne I, Scott S, Figg N, Kirkpatrick P, Ritchie A, Goddard M, Bennett M . Vascular smooth muscle cells undergo telomere-based senescence in human atherosclerosis: effects of telomerase and oxidative stress. Circ Res. 2006; 99: 156–164.LinkGoogle Scholar
    • 24. O'Sullivan M, Scott SD, McCarthy N, Figg N, Shapiro LM, Kirkpatrick P, Bennett MR . Differential cyclin E expression in human in-stent stenosis smooth muscle cells identifies targets for selective anti-restenosis therapy. Cardiovasc Res. 2003; 60: 673–683.CrossrefMedlineGoogle Scholar
    • 25. Bennett MR, Macdonald K, Chan SW, Boyle JJ, Weissberg PL . Cooperative interactions between RB and p53 regulate cell proliferation, cell senescence, and apoptosis in human vascular smooth muscle cells from atherosclerotic plaques. Circ Res. 1998; 82: 704–712.LinkGoogle Scholar
    • 26. Edelberg JM, Tang L, Hattori K, Lyden D, Rafii S . Young adult bone marrow-derived endothelial precursor cells restore aging-impaired cardiac angiogenic function. Circ Res. 2002; 90: E89–E93.LinkGoogle Scholar
    • 27. Xu Q, Wang J, He J, Zhou M, Adi J, Webster KA, Yu H . Impaired CXCR4 expression and cell engraftment of bone marrow-derived cells from aged atherogenic mice. Atherosclerosis. 2011; 219: 92–99.CrossrefMedlineGoogle Scholar
    • 28. Bentzon JF, Weile C, Sondergaard CS, Hindkjaer J, Kassem M, Falk E . Smooth muscle cells in atherosclerosis originate from the local vessel wall and not circulating progenitor cells in ApoE knockout mice. Arterioscler Thromb Vasc Biol. 2006; 26: 2696–2702.LinkGoogle Scholar
    • 29. Hagensen MK, Raarup MK, Mortensen MB, Thim T, Nyengaard JR, Falk E, Bentzon JF . Circulating endothelial progenitor cells do not contribute to regeneration of endothelium after murine arterial injury. Cardiovasc Res. 2012; 93: 223–231.CrossrefMedlineGoogle Scholar
    • 30. Hagensen MK, Shim J, Thim T, Falk E, Bentzon JF . Circulating endothelial progenitor cells do not contribute to plaque endothelium in murine atherosclerosis. Circulation. 2010; 121: 898–905.LinkGoogle Scholar
    • 31. Heiss C, Keymel S, Niesler U, Ziemann J, Kelm M, Kalka C . Impaired progenitor cell activity in age-related endothelial dysfunction. J Am Coll Cardiol. 2005; 45: 1441–1448.CrossrefMedlineGoogle Scholar
    • 32. Shao H, Xu Q, Wu Q, Ma Q, Salgueiro L, Wang J, Eton D, Webster KA, Yu H . Defective CXCR4 expression in aged bone marrow cells impairs vascular regeneration. J Cell Mol Med. 2011; 15: 2046–2056.CrossrefMedlineGoogle Scholar
    • 33. Bosch-Marce M, Okuyama H, Wesley JB , et al . Effects of aging and hypoxia-inducible factor-1 activity on angiogenic cell mobilization and recovery of perfusion after limb ischemia. Circ Res. 2007; 101: 1310–1318.LinkGoogle Scholar
    • 34. Chang EI, Loh SA, Ceradini DJ, Chang EI, Lin SE, Bastidas N, Aarabi S, Chan DA, Freedman ML, Giaccia AJ, Gurtner GC . Age decreases endothelial progenitor cell recruitment through decreases in hypoxia-inducible factor 1alpha stabilization during ischemia. Circulation. 2007; 116: 2818–2829.LinkGoogle Scholar
    • 35. Zhang Y, Herbert BS, Rajashekhar G, Ingram DA, Yoder MC, Clauss M, Rehman J . Premature senescence of highly proliferative endothelial progenitor cells is induced by tumor necrosis factor-alpha via the p38 mitogen-activated protein kinase pathway. FASEB J. 2009; 23: 1358–1365.CrossrefMedlineGoogle Scholar
    • 36. Carracedo J, Merino A, Briceno C, Soriano S, Buendia P, Calleros L, Rodriguez M, Martin-Malo A, Aljama P, Ramirez R . Carbamylated low-density lipoprotein induces oxidative stress and accelerated senescence in human endothelial progenitor cells. FASEB J. 2011; 25: 1314–1322.CrossrefMedlineGoogle Scholar
    • 37. Dimri G, Lee X, Basile G, Acosta M, Scott G, Roskelley C, Medranos E, Linskens M, Rubelj I, Pereira-Smith O, Peacocke M . A biomarker that identifies senescent human cells in culture and in aging skin in vivo. Proc Natl Acad Sci U S A. 1995; 92: 9363–9367.CrossrefMedlineGoogle Scholar
    • 38. 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: 1541–1544.LinkGoogle Scholar
    • 39. Okuda K, Bardeguez A, Gardner JP, Rodriguez P, Ganesh V, Kimura M, Skurnick J, Awad G, Aviv A . Telomere length in the newborn. Pediatr Res. 2002; 52: 377–381.CrossrefMedlineGoogle Scholar
    • 40. Sahin E, Colla S, Liesa M , et al . Telomere dysfunction induces metabolic and mitochondrial compromise. Nature. 2011; 470: 359–365.CrossrefMedlineGoogle Scholar
    • 41. de Lange T . Shelterin: the protein complex that shapes and safeguards human telomeres. Genes Dev. 2005; 19: 2100–2110.CrossrefMedlineGoogle Scholar
    • 42. Zeng S, Xiang T, Pandita TK, Gonzalez-Suarez I, Gonzalo S, Harris CC, Yang Q . Telomere recombination requires the MUS81 endonuclease. Nat Cell Biol. 2009; 11: 616–623.CrossrefMedlineGoogle Scholar
    • 43. Kim H, Lee OH, Xin H, Chen LY, Qin J, Chae HK, Lin SY, Safari A, Liu D, Songyang Z . TRF2 functions as a protein hub and regulates telomere maintenance by recognizing specific peptide motifs. Nat Struct Mol Biol. 2009; 16: 372–379.CrossrefMedlineGoogle Scholar
    • 44. Smogorzewska A, van Steensel B, Bianchi A, Oelmann S, Schaefer MR, Schnapp G, de Lange T . Control of human telomere length by TRF1 and TRF2. Mol Cell Biol. 2000; 20: 1659–1668.CrossrefMedlineGoogle Scholar
    • 45. Wang F, Podell ER, Zaug AJ, Yang Y, Baciu P, Cech TR, Lei M . The POT1-TPP1 telomere complex is a telomerase processivity factor. Nature. 2007; 445: 506–510.CrossrefMedlineGoogle Scholar
    • 46. Etheridge KT, Compton SA, Barrientos KS, Ozgur S, Griffith JD, Counter CM . Tethering telomeric double- and single-stranded DNA-binding proteins inhibits telomere elongation. J Biol Chem. 2008; 283: 6935–6941.CrossrefMedlineGoogle Scholar
    • 47. Karlseder J, Smogorzewska A, de Lange T . Senescence induced by altered telomere state, not telomere loss. Science. 2002; 295: 2446–2449.CrossrefMedlineGoogle Scholar
    • 48. van Steensel B, Smogorzewska A, de Lange T . TRF2 protects human telomeres from end-to-end fusions. Cell. 1998; 92: 401–413.CrossrefMedlineGoogle Scholar
    • 49. Karlseder J, Broccoli D, Dai Y, Hardy S, de Lange T . p53- and ATM-dependent apoptosis induced by telomeres lacking TRF2. Science. 1999; 283: 1321–1325.CrossrefMedlineGoogle Scholar
    • 50. Wang RC, Smogorzewska A, de Lange T . Homologous recombination generates T-loop-sized deletions at human telomeres. Cell. 2004; 119: 355–368.CrossrefMedlineGoogle Scholar
    • 51. Ogami M, Ikura Y, Ohsawa M, Matsuo T, Kayo S, Yoshimi N, Hai E, Shirai N, Ehara S, Komatsu R, Naruko T, Ueda M . Telomere shortening in human coronary artery diseases. Arterioscler Thromb Vasc Biol. 2004; 24: 546–550.LinkGoogle Scholar
    • 52. Brouilette S, Singh RK, Thompson JR, Goodall AH, Samani NJ . White cell telomere length and risk of premature myocardial infarction. Arterioscler Thromb Vasc Biol. 2003; 23: 842–846.LinkGoogle Scholar
    • 53. Brouilette SW, Moore JS, McMahon AD, Thompson JR, Ford I, Shepherd J, Packard CJ, Samani NJ . Telomere length, risk of coronary heart disease, and statin treatment in the West of Scotland Primary Prevention Study: a nested case-control study. Lancet. 2007; 369: 107–114.CrossrefMedlineGoogle Scholar
    • 54. Panayiotou AG, Nicolaides AN, Griffin M, Tyllis T, Georgiou N, Bond D, Martin RM, Hoppensteadt D, Fareed J, Humphries SE . Leukocyte telomere length is associated with measures of subclinical atherosclerosis. Atherosclerosis. 2010; 211: 176–181.CrossrefMedlineGoogle Scholar
    • 55. Willeit P, Willeit J, Brandstatter A, Ehrlenbach S, Mayr A, Gasperi A, Weger S, Oberhollenzer F, Reindl M, Kronenberg F, Kiechl S . Cellular aging reflected by leukocyte telomere length predicts advanced atherosclerosis and cardiovascular disease risk. Arterioscler Thromb Vasc Biol. 2010; 30: 1649–1656.LinkGoogle Scholar
    • 56. Chang E, Harley CB . Telomere length and replicative aging in human vascular tissues. Proc Natl Acad Sci U S A. 1995; 92: 11190–11194.CrossrefMedlineGoogle Scholar
    • 57. Nzietchueng R, Elfarra M, Nloga J, Labat C, Carteaux JP, Maureira P, Lacolley P, Villemot JP, Benetos A . Telomere length in vascular tissues from patients with atherosclerotic disease. J Nutr Health Aging. 2011; 15: 153–156.CrossrefMedlineGoogle Scholar
    • 58. Botto N, Rizza A, Colombo MG, Mazzone AM, Manfredi S, Masetti S, Clerico A, Biagini A, Andreassi MG . Evidence for DNA damage in patients with coronary artery disease. Mutat Res. 2001; 493: 23–30.CrossrefMedlineGoogle Scholar
    • 59. Martinet W, Knaapen MW, De Meyer GR, Herman AG, Kockx MM . Elevated levels of oxidative DNA damage and DNA repair enzymes in human atherosclerotic plaques. Circulation. 2002; 106: 927–932.LinkGoogle Scholar
    • 60. Botto N, Berti S, Manfredi S, Al-Jabri A, Federici C, Clerico A, Ciofini E, Biagini A, Andreassi MG . Detection of mtDNA with 4977 bp deletion in blood cells and atherosclerotic lesions of patients with coronary artery disease. Mutat Res. 2005; 570: 81–88.CrossrefMedlineGoogle Scholar
    • 61. Mahmoudi M, Gorenne I, Mercer J, Figg N, Littlewood T, Bennett M . Statins use a novel Nijmegen breakage syndrome-1-dependent pathway to accelerate DNA repair in vascular smooth muscle cells. Circ Res. 2008; 103: 717–725.LinkGoogle Scholar
    • 62. Gray K, Bennett M . Role of DNA damage in atherosclerosis: bystander or participant?Biochem Pharmacol. 2011; 82: 693–700.CrossrefMedlineGoogle Scholar
    • 63. Mercer JR, Cheng KK, Figg N, Gorenne I, Mahmoudi M, Griffin J, Vidal-Puig A, Logan A, Murphy MP, Bennett M . DNA damage links mitochondrial dysfunction to atherosclerosis and the metabolic syndrome. Circ Res. 2011; 107: 1021–1031.LinkGoogle Scholar
    • 64. Fujimoto H, Kobayashi H, Ohno M . Age-induced reduction in mitochondrial manganese superoxide dismutase activity and tolerance of macrophages against apoptosis induced by oxidized low density lipoprotein. Circ J. 2010; 74: 353–360.CrossrefMedlineGoogle Scholar
    • 65. Tatarkova Z, Kuka S, Racay P, Lehotsky J, Dobrota D, Mistuna D, Kaplan P . Effects of aging on activities of mitochondrial electron transport chain complexes and oxidative damage in rat heart. Physiol Res. 2011; 60: 281–289.CrossrefMedlineGoogle Scholar
    • 66. Bandyopadhyay D, Medrano EE . The emerging role of epigenetics in cellular and organismal aging. Exp Gerontol. 2003; 38: 1299–1307.CrossrefMedlineGoogle Scholar
    • 67. Calvanese V, Lara E, Kahn A, Fraga MF . The role of epigenetics in aging and age-related diseases. Ageing Res Rev. 2009; 8: 268–276.CrossrefMedlineGoogle Scholar
    • 68. Lund G, Zaina S . Atherosclerosis: an epigenetic balancing act that goes wrong. Curr Atheroscler Rep. 2011; 13: 208–214.CrossrefMedlineGoogle Scholar
    • 69. Zhang FF, Cardarelli R, Carroll J, Fulda KG, Kaur M, Gonzalez K, Vishwanatha JK, Santella RM, Morabia A . Significant differences in global genomic DNA methylation by gender and race/ethnicity in peripheral blood. Epigenetics. 2011; 6: 623–629.CrossrefMedlineGoogle Scholar
    • 70. Madrigano J, Baccarelli A, Mittleman MA, Sparrow D, Vokonas PS, Tarantini L, Schwartz J . Aging and epigenetics: longitudinal changes in gene-specific DNA methylation. Epigenetics. 2012; 7: 63–70.CrossrefMedlineGoogle Scholar
    • 71. Hiltunen MO, Turunen MP, Hakkinen TP, Rutanen J, Hedman M, Makinen K, Turunen AM, Aalto-Setala K, Yla-Herttuala S . DNA hypomethylation and methyltransferase expression in atherosclerotic lesions. Vasc Med. 2002; 7: 5–11.CrossrefMedlineGoogle Scholar
    • 72. Castillo-Diaz SA, Garay-Sevilla ME, Hernandez-Gonzalez MA, Solis-Martinez MO, Zaina S . Extensive demethylation of normally hypermethylated CpG islands occurs in human atherosclerotic arteries. Int J Mol Med. 2010; 26: 691–700.MedlineGoogle Scholar
    • 73. Kim M, Long TI, Arakawa K, Wang R, Yu MC, Laird PW . DNA methylation as a biomarker for cardiovascular disease risk. PLoS One. 2010; 5: e9692.CrossrefMedlineGoogle Scholar
    • 74. Lund G, Andersson L, Lauria M, Lindholm M, Fraga MF, Villar-Garea A, Ballestar E, Esteller M, Zaina S . DNA methylation polymorphisms precede any histological sign of atherosclerosis in mice lacking apolipoprotein E. J Biol Chem. 2004; 279: 29147–29154.CrossrefMedlineGoogle Scholar
    • 75. Ventura A, Luzi L, Pacini S, Baldari CT, Pelicci PG . The p66Shc longevity gene is silenced through epigenetic modifications of an alternative promoter. J Biol Chem. 2002; 277: 22370–22376.CrossrefMedlineGoogle Scholar
    • 76. Napoli C, Martin-Padura I, de Nigris F, Giorgio M, Mansueto G, Somma P, Condorelli M, Sica G, De Rosa G, Pelicci P . Deletion of the p66Shc longevity gene reduces systemic and tissue oxidative stress, vascular cell apoptosis, and early atherogenesis in mice fed a high-fat diet. Proc Natl Acad Sci U S A. 2003; 100: 2112–2116.CrossrefMedlineGoogle Scholar
    • 77. Francia P, delli Gatti C, Bachschmid M, Martin-Padura I, Savoia C, Migliaccio E, Pelicci PG, Schiavoni M, Luscher TF, Volpe M, Cosentino F . Deletion of p66shc gene protects against age-related endothelial dysfunction. Circulation. 2004; 110: 2889–2895.LinkGoogle Scholar
    • 78. Spyridopoulos I, Erben Y, Brummendorf T, Haendeler J, Dietz K, Seeger F, Kissel C, Martin H, Hoffmann J, Assmus B, Zeiher A, Dimmeler S . Telomere gap between granulocytes and lymphocytes is a determinant for hematopoetic progenitor cell impairment in patients with previous myocardial infarction. Arterioscler Thromb Vasc Biol. 2008; 28: 968–974.LinkGoogle Scholar
    • 79. Jaskelioff M, Muller FL, Paik JH, Thomas E, Jiang S, Adams AC, Sahin E, Kost-Alimova M, Protopopov A, Cadinanos J, Horner JW, Maratos-Flier E, Depinho RA . Telomerase reactivation reverses tissue degeneration in aged telomerase-deficient mice. Nature. 2011; 469: 102–106.CrossrefMedlineGoogle Scholar
    • 80. Poch E, Carbonell P, Franco S, Diez-Juan A, Blasco MA, Andres V . Short telomeres protect from diet-induced atherosclerosis in apolipoprotein E-null mice. FASEB J. 2004; 18: 418–420.CrossrefMedlineGoogle Scholar
    • 81. Calvert PA, Liew TV, Gorenne I , et al . Leukocyte telomere length is associated with high-risk plaques on virtual histology intravascular ultrasound and increased proinflammatory activity. Arterioscler Thromb Vasc Biol. 2011; 31: 2157–2164.LinkGoogle Scholar
    • 82. Herbert KE, Mistry Y, Hastings R, Poolman T, Niklason L, Williams B . Angiotensin II-mediated oxidative DNA damage accelerates cellular senescence in cultured human vascular smooth muscle cells via telomere-dependent and independent pathways. Circ Res. 2008; 102: 201–208.LinkGoogle Scholar
    • 83. Voghel G, Thorin-Trescases N, Mamarbachi AM, Villeneuve L, Mallette FA, Ferbeyre G, Farhat N, Perrault LP, Carrier M, Thorin E . Endogenous oxidative stress prevents telomerase-dependent immortalization of human endothelial cells. Mech Ageing Dev. 2010; 131: 354–363.CrossrefMedlineGoogle Scholar
    • 84. Schneider JG, Finck BN, Ren J, Standley KN, Takagi M, Maclean KH, Bernal-Mizrachi C, Muslin AJ, Kastan MB, Semenkovich CF . ATM-dependent suppression of stress signaling reduces vascular disease in metabolic syndrome. Cell Metab. 2006; 4: 377–389.CrossrefMedlineGoogle Scholar
    • 85. Ragnauth CD, Warren DT, Liu Y, McNair R, Tajsic T, Figg N, Shroff R, Skepper J, Shanahan CM . Prelamin A acts to accelerate smooth muscle cell senescence and is a novel biomarker of human vascular aging. Circulation. 2010; 121: 2200–2210.LinkGoogle Scholar
    • 86. Liu B, Wang J, Chan KM , et al . Genomic instability in laminopathy-based premature aging. Nat Med. 2005; 11: 780–785.CrossrefMedlineGoogle Scholar
    • 87. McClintock D, Gordon LB, Djabali K . Hutchinson-Gilford progeria mutant lamin A primarily targets human vascular cells as detected by an anti-Lamin A G608G antibody. Proc Natl Acad Sci U S A. 2006; 103: 2154–2159.CrossrefMedlineGoogle Scholar
    • 88. Nehlin JO, Skovgaard GL, Bohr VA . The Werner syndrome: a model for the study of human aging. Ann N Y Acad Sci. 2000; 908: 167–179.CrossrefMedlineGoogle Scholar
    • 89. Lebel M . Werner syndrome: genetic and molecular basis of a premature aging disorder. Cell Mol Life Sci. 2001; 58: 857–867.CrossrefMedlineGoogle Scholar
    • 90. Moon SK, Thompson LJ, Madamanchi N, Ballinger S, Papaconstantinou J, Horaist C, Runge MS, Patterson C . Aging, oxidative responses, and proliferative capacity in cultured mouse aortic smooth muscle cells. Am J Physiol Heart Circ Physiol. 2001; 280: H2779–H2788.CrossrefMedlineGoogle Scholar
    • 91. Guntani A, Matsumoto T, Kyuragi R, Iwasa K, Onohara T, Itoh H, Katusic ZS, Maehara Y . Reduced proliferation of aged human vascular smooth muscle cells: role of oxygen-derived free radicals and BubR1 expression. J Surg Res. 2011; 170: 143–149.CrossrefMedlineGoogle Scholar
    • 92. Mowbray AL, Kang DH, Rhee SG, Kang SW, Jo H . Laminar shear stress up-regulates peroxiredoxins (PRX) in endothelial cells: PRX 1 as a mechanosensitive antioxidant. J Biol Chem. 2008; 283: 1622–1627.CrossrefMedlineGoogle Scholar
    • 93. Nihei S, Yamashita K, Tasaki H, Ozumi K, Nakashima Y . Oxidized low-density lipoprotein-induced apoptosis is attenuated by insulin-activated phosphatidylinositol 3-kinase/Akt through p38 mitogen-activated protein kinase. Clin Exp Pharmacol Physiol. 2005; 32: 224–229.CrossrefMedlineGoogle Scholar
    • 94. Breitschopf K, Zeiher AM, Dimmeler S . Pro-atherogenic factors induce telomerase inactivation in endothelial cells through an Akt-dependent mechanism. FEBS Lett. 2001; 493: 21–25.CrossrefMedlineGoogle Scholar
    • 95. Kavurma MM, Figg N, Bennett MR, Mercer J, Khachigian LM, Littlewood TD . Oxidative stress regulates IGF1R expression in vascular smooth-muscle cells via p53 and HDAC recruitment. Biochem J. 2007; 407: 79–87.CrossrefMedlineGoogle Scholar
    • 96. Barcellos-Hoff MH, Dix TA . Redox-mediated activation of latent transforming growth factor-beta 1. Mol Endocrinol. 1996; 10: 1077–1083.MedlineGoogle Scholar
    • 97. Fuhrman B, Volkova N, Aviram M . Oxidative stress increases the expression of the CD36 scavenger receptor and the cellular uptake of oxidized low-density lipoprotein in macrophages from atherosclerotic mice: protective role of antioxidants and of paraoxonase. Atherosclerosis. 2002; 161: 307–316.CrossrefMedlineGoogle Scholar
    • 98. Zhu S, Goldschmidt-Clermont PJ, Dong C . Inactivation of monocarboxylate transporter MCT3 by DNA methylation in atherosclerosis. Circulation. 2005; 112: 1353–1361.LinkGoogle Scholar
    • 99. Nakamura Y, Suzuki T, Miki Y, Tazawa C, Senzaki K, Moriya T, Saito H, Ishibashi T, Takahashi S, Yamada S, Sasano H . Estrogen receptors in atherosclerotic human aorta: inhibition of human vascular smooth muscle cell proliferation by estrogens. Mol Cell Endocrinol. 2004; 219: 17–26.CrossrefMedlineGoogle Scholar
    • 100. Post WS, Goldschmidt-Clermont PJ, Wilhide CC, Heldman AW, Sussman MS, Ouyang P, Milliken EE, Issa JP . Methylation of the estrogen receptor gene is associated with aging and atherosclerosis in the cardiovascular system. Cardiovasc Res. 1999; 43: 985–991.CrossrefMedlineGoogle Scholar
    • 101. Kim J, Kim JY, Song KS, Lee YH, Seo JS, Jelinek J, Goldschmidt-Clermont PJ, Issa JP . Epigenetic changes in estrogen receptor beta gene in atherosclerotic cardiovascular tissues and in-vitro vascular senescence. Biochim Biophys Acta. 2007; 1772: 72–80.CrossrefMedlineGoogle Scholar
    • 102. Ying AK, Hassanain HH, Roos CM, Smiraglia DJ, Issa JJ, Michler RE, Caligiuri M, Plass C, Goldschmidt-Clermont PJ . Methylation of the estrogen receptor-alpha gene promoter is selectively increased in proliferating human aortic smooth muscle cells. Cardiovasc Res. 2000; 46: 172–179.CrossrefMedlineGoogle Scholar
    • 103. Ji R, Cheng Y, Yue J, Yang J, Liu X, Chen H, Dean DB, Zhang C . MicroRNA expression signature and antisense-mediated depletion reveal an essential role of MicroRNA in vascular neointimal lesion formation. Circ Res. 2007; 100: 1579–1588.LinkGoogle Scholar
    • 104. Minami Y, Satoh M, Maesawa C, Takahashi Y, Tabuchi T, Itoh T, Nakamura M . Effect of atorvastatin on microRNA 221/222 expression in endothelial progenitor cells obtained from patients with coronary artery disease. Eur J Clin Invest. 2009; 39: 359–367.CrossrefMedlineGoogle Scholar
    • 105. Liu X, Cheng Y, Zhang S, Lin Y, Yang J, Zhang C . A necessary role of miR-221 and miR-222 in vascular smooth muscle cell proliferation and neointimal hyperplasia. Circ Res. 2009; 104: 476–487.LinkGoogle Scholar
    • 106. Davis BN, Hilyard AC, Nguyen PH, Lagna G, Hata A . Induction of microRNA-221 by platelet-derived growth factor signaling is critical for modulation of vascular smooth muscle phenotype. J Biol Chem. 2009; 284: 3728–3738.CrossrefMedlineGoogle Scholar
    • 107. Davis-Dusenbery BN, Chan MC, Reno KE, Weisman AS, Layne MD, Lagna G, Hata A . Down-regulation of Kruppel-like factor-4 (KLF4) by microRNA-143/145 is critical for modulation of vascular smooth muscle cell phenotype by transforming growth factor-beta and bone morphogenetic protein 4. J Biol Chem. 2011; 286: 28097–28110.CrossrefMedlineGoogle Scholar
    • 108. Cheng Y, Liu X, Yang J, Lin Y, Xu DZ, Lu Q, Deitch EA, Huo Y, Delphin ES, Zhang C . MicroRNA-145, a novel smooth muscle cell phenotypic marker and modulator, controls vascular neointimal lesion formation. Circ Res. 2009; 105: 158–166.LinkGoogle Scholar
    • 109. Cordes KR, Sheehy NT, White MP, Berry EC, Morton SU, Muth AN, Lee TH, Miano JM, Ivey KN, Srivastava D . miR-145 and miR-143 regulate smooth muscle cell fate and plasticity. Nature. 2009; 460: 705–710.CrossrefMedlineGoogle Scholar
    • 110. Rippe C, Blimline M, Magerko KA, Lawson BR, LaRocca TJ, Donato AJ, Seals DR . MicroRNA changes in human arterial endothelial cells with senescence: relation to apoptosis, eNOS and inflammation. Exp Gerontol. 47: 45–51.CrossrefMedlineGoogle Scholar
    • 111. Rayner KJ, Esau CC, Hussain FN , et al . Inhibition of miR-33a/b in non-human primates raises plasma HDL and lowers VLDL triglycerides. Nature. 2011; 478: 404–407.CrossrefMedlineGoogle Scholar
    • 112. Davalos A, Goedeke L, Smibert P , et al . miR-33a/b contribute to the regulation of fatty acid metabolism and insulin signaling. Proc Natl Acad Sci U S A. 2011; 108: 9232–9237.CrossrefMedlineGoogle Scholar
    • 113. Chaturvedi N . Ethnic differences in cardiovascular disease. Heart. 2003; 89: 681–686.CrossrefMedlineGoogle Scholar
    • 114. Chaturvedi N, Bulpitt CJ, Leggetter S, Schiff R, Nihoyannopoulos P, Strain WD, Shore AC, Rajkumar C . Ethnic differences in vascular stiffness and relations to hypertensive target organ damage. J Hypertens. 2004; 22: 1731–1737.CrossrefMedlineGoogle Scholar
    • 115. Kalra L, Rambaran C, Chowienczyk P, Goss D, Hambleton I, Ritter J, Shah A, Wilks R, Forrester T . Ethnic differences in arterial responses and inflammatory markers in Afro-Caribbean and Caucasian subjects. Arterioscler Thromb Vasc Biol. 2005; 25: 2362–2367.LinkGoogle Scholar
    • 116. Smulyan H, Asmar RG, Rudnicki A, London GM, Safar ME . Comparative effects of aging in men and women on the properties of the arterial tree. J Am Coll Cardiol. 2001; 37: 1374–1380.CrossrefMedlineGoogle Scholar
    • 117. Ahimastos AA, Formosa M, Dart AM, Kingwell BA . Gender differences in large artery stiffness pre- and post puberty. J Clin Endocrinol Metab. 2003; 88: 5375–5380.CrossrefMedlineGoogle Scholar
    • 118. Rajkumar C, Kingwell BA, Cameron JD, Waddell T, Mehra R, Christophidis N, Komesaroff PA, McGrath B, Jennings GL, Sudhir K, Dart AM . Hormonal therapy increases arterial compliance in postmenopausal women. J Am Coll Cardiol. 1997; 30: 350–356.CrossrefMedlineGoogle Scholar
    • 119. Dockery F, Bulpitt CJ, Donaldson M, Fernandez S, Rajkumar C . The relationship between androgens and arterial stiffness in older men. J Am Geriatr Soc. 2003; 51: 1627–1632.CrossrefMedlineGoogle Scholar
    • 120. Waddell TK, Rajkumar C, Cameron JD, Jennings GL, Dart AM, Kingwell BA . Withdrawal of hormonal therapy for 4 weeks decreases arterial compliance in postmenopausal women. J Hypertens. 1999; 17: 413–418.CrossrefMedlineGoogle Scholar
    • 121. Mahmud A, Feely J . Effect of smoking on arterial stiffness and pulse pressure amplification. Hypertension. 2003; 41: 183–187.LinkGoogle Scholar
    • 122. Nyunoya T, Monick MM, Klingelhutz AL, Glaser H, Cagley JR, Brown CO, Matsumoto E, Aykin-Burns N, Spitz DR, Oshima J, Hunninghake GW . Cigarette smoke induces cellular senescence via Werner's syndrome protein down-regulation. Am J Respir Crit Care Med. 2009; 179: 279–287.CrossrefMedlineGoogle Scholar
    • 123. Sierksma A, Muller M, van der Schouw YT, Grobbee DE, Hendriks HF, Bots ML . Alcohol consumption and arterial stiffness in men. J Hypertens. 2004; 22: 357–362.CrossrefMedlineGoogle Scholar
    • 124. Mahmud A, Feely J . Divergent effect of acute and chronic alcohol on arterial stiffness. Am J Hypertens. 2002; 15: 240–243.CrossrefMedlineGoogle Scholar
    • 125. Stacey RB, Bertoni AG, Eng J, Bluemke DA, Hundley WG, Herrington D . Modification of the effect of glycemic status on aortic distensibility by age in the multi-ethnic study of atherosclerosis. Hypertension. 2010; 55: 26–32.LinkGoogle Scholar
    • 126. Matsui-Hirai H, Hayashi T, Yamamoto S, Ina K, Maeda M, Kotani H, Iguchi A, Ignarro LJ, Hattori Y . Dose-dependent modulatory effects of insulin on glucose-induced endothelial senescence in vitro and in vivo: a relationship between telomeres and nitric oxide. J Pharmacol Exp Ther. 2011; 337: 591–599.CrossrefMedlineGoogle Scholar
    • 127. Oimomi M, Igaki N, Hata F, Kitamura Y, Nishimoto S, Baba S, Maeda S . Age- and diabetes-accelerated glycation in the human aorta. Arch Gerontol Geriatr. 1989; 8: 123–127.CrossrefMedlineGoogle Scholar
    • 128. Vlassara H, Fuh H, Makita Z, Krungkrai S, Cerami A, Bucala R . Exogenous advanced glycosylation end products induce complex vascular dysfunction in normal animals: a model for diabetic and aging complications. Proc Natl Acad Sci U S A. 1992; 89: 12043–12047.CrossrefMedlineGoogle Scholar
    • 129. Sakata N, Meng J, Jimi S, Takebayashi S . Nonenzymatic glycation and extractability of collagen in human atherosclerotic plaques. Atherosclerosis. 1995; 116: 63–75.CrossrefMedlineGoogle Scholar
    • 130. Rojas A, Romay S, Gonzalez D, Herrera B, Delgado R, Otero K . Regulation of endothelial nitric oxide synthase expression by albumin-derived advanced glycosylation end products. Circ Res. 2000; 86: E50–E54.LinkGoogle Scholar
    • 131. Schmidt AM, Hori O, Brett J, Yan SD, Wautier JL, Stern D . Cellular receptors for advanced glycation end products: implications for induction of oxidant stress and cellular dysfunction in the pathogenesis of vascular lesions. Arterioscler Thromb. 1994; 14: 1521–1528.LinkGoogle Scholar
    • 132. Vlassara H, Fuh H, Donnelly T, Cybulsky M . Advanced glycation endproducts promote adhesion molecule (VCAM-1, ICAM-1) expression and atheroma formation in normal rabbits. Mol Med. 1995; 1: 447–456.CrossrefMedlineGoogle Scholar
    • 133. Higashi T, Sano H, Saishoji T, Ikeda K, Jinnouchi Y, Kanzaki T, Morisaki N, Rauvala H, Shichiri M, Horiuchi S . The receptor for advanced glycation end products mediates the chemotaxis of rabbit smooth muscle cells. Diabetes. 1997; 46: 463–472.CrossrefMedlineGoogle Scholar
    • 134. Jinnouchi Y, Sano H, Nagai R, Hakamata H, Kodama T, Suzuki H, Yoshida M, Ueda S, Horiuchi S . Glycolaldehyde-modified low density lipoprotein leads macrophages to foam cells via the macrophage scavenger receptor. J Biochem. 1998; 123: 1208–1217.CrossrefMedlineGoogle Scholar
    • 135. Yan SD, Schmidt AM, Anderson GM, Zhang J, Brett J, Zou YS, Pinsky D, Stern D . Enhanced cellular oxidant stress by the interaction of advanced glycation end products with their receptors/binding proteins. J Biol Chem. 1994; 269: 9889–9897.CrossrefMedlineGoogle Scholar
    • 136. Sano H, Nagai R, Matsumoto K, Horiuchi S . Receptors for proteins modified by advanced glycation endproducts (AGE): their functional role in atherosclerosis. Mech Ageing Dev. 1999; 107: 333–346.CrossrefMedlineGoogle Scholar
    • 137. Lin RY, Reis ED, Dore AT, Lu M, Ghodsi N, Fallon JT, Fisher EA, Vlassara H . Lowering of dietary advanced glycation endproducts (AGE) reduces neointimal formation after arterial injury in genetically hypercholesterolemic mice. Atherosclerosis. 2002; 163: 303–311.CrossrefMedlineGoogle Scholar
    • 138. Lin RY, Choudhury RP, Cai W, Lu M, Fallon JT, Fisher EA, Vlassara H . Dietary glycotoxins promote diabetic atherosclerosis in apolipoprotein E-deficient mice. Atherosclerosis. 2003; 168: 213–220.CrossrefMedlineGoogle Scholar
    • 139. Nicholl ID, Stitt AW, Moore JE, Ritchie AJ, Archer DB, Bucala R . Increased levels of advanced glycation endproducts in the lenses and blood vessels of cigarette smokers. Mol Med. 1998; 4: 594–601.CrossrefMedlineGoogle Scholar
    • 140. Wagner M, Hampel B, Bernhard D, Hala M, Zwerschke W, Jansen-Durr P . Replicative senescence of human endothelial cells in vitro involves G1 arrest, polyploidization and senescence-associated apoptosis. Exp Gerontol. 2001; 36: 1327–1347.CrossrefMedlineGoogle Scholar
    • 141. Asai K, Kudej RK, Shen YT, Yang GP, Takagi G, Kudej AB, Geng YJ, Sato N, Nazareno JB, Vatner DE, Natividad F, Bishop SP, Vatner SF . Peripheral vascular endothelial dysfunction and apoptosis in old monkeys. Arterioscler Thromb Vasc Biol. 2000; 20: 1493–1499.LinkGoogle Scholar
    • 142. Shi Q, Aida K, Vandeberg JL, Wang XL . Passage-dependent changes in baboon endothelial cells–relevance to in vitro aging. DNA Cell Biol. 2004; 23: 502–509.CrossrefMedlineGoogle Scholar
    • 143. Khaidakov M, Wang X, Mehta JL . Potential involvement of LOX-1 in functional consequences of endothelial senescence. PLoS One. 2011; 6: e20964.CrossrefMedlineGoogle Scholar
    • 144. 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. Biochem Biophys Res Commun. 1993; 195: 1070–1076.CrossrefMedlineGoogle Scholar
    • 145. Donato AJ, Gano LB, Eskurza I, Silver AE, Gates PE, Jablonski K, Seals DR . Vascular endothelial dysfunction with aging: endothelin-1 and endothelial nitric oxide synthase. Am J Physiol Heart Circ Physiol. 2009; 297: H425–H432.CrossrefMedlineGoogle Scholar
    • 146. Zhou X, Perez F, Han K, Jurivich DA . Clonal senescence alters endothelial ICAM-1 function. Mech Ageing Dev. 2006; 127: 779–785.CrossrefMedlineGoogle Scholar
    • 147. Song Y, Shen H, Schenten D, Shan P, Lee PJ, Goldstein DR . Aging enhances the basal production of IL-6 and CCL2 in vascular smooth muscle cells. Arterioscler Thromb Vasc Biol. 2012; 32: 103–109.LinkGoogle Scholar
    • 148. Razani B, Feng C, Semenkovich CF . p53 is required for chloroquine-induced atheroprotection but not insulin sensitization. J Lipid Res. 2010; 51: 1738–1746.CrossrefMedlineGoogle Scholar
    • 149. Werner C, Gensch C, Poss J, Haendeler J, Bohm M, Laufs U . Pioglitazone activates aortic telomerase and prevents stress-induced endothelial apoptosis. Atherosclerosis. 2011; 216: 23–34.CrossrefMedlineGoogle Scholar
    • 150. Harley CB . Telomerase and cancer therapeutics. Nat Rev Cancer. 2008; 8: 167–179.CrossrefMedlineGoogle Scholar
    • 151. t Hoen PA, Van der Lans CA, Van Eck M, Bijsterbosch MK, Van Berkel TJ, Twisk J . Aorta of ApoE-deficient mice responds to atherogenic stimuli by a prelesional increase and subsequent decrease in the expression of antioxidant enzymes. Circ Res. 2003; 93: 262–269.LinkGoogle Scholar
    • 152. Collins AR, Lyon CJ, Xia X, Liu JZ, Tangirala RK, Yin F, Boyadjian R, Bikineyeva A, Pratico D, Harrison DG, Hsueh WA . Age-accelerated atherosclerosis correlates with failure to upregulate antioxidant genes. Circ Res. 2009; 104: e42–e54.LinkGoogle Scholar
    • 153. Ricciarelli R, Zingg JM, Azzi A . Vitamin E reduces the uptake of oxidized LDL by inhibiting CD36 scavenger receptor expression in cultured aortic smooth muscle cells. Circulation. 2000; 102: 82–87.LinkGoogle Scholar
    • 154. Park LK, Friso S, Choi SW . Nutritional influences on epigenetics and age-related disease. Proc Nutr Soc. 2012; 71: 75–83.CrossrefMedlineGoogle Scholar
    • 155. Yusuf S, Dagenais G, Pogue J, Bosch J, Sleight P . Vitamin E supplementation and cardiovascular events in high-risk patients: the Heart Outcomes Prevention Evaluation Study Investigators. N Engl J Med. 2000; 342: 154–160.CrossrefMedlineGoogle Scholar
    • 156. Cook NR, Albert CM, Gaziano JM, Zaharris E, MacFadyen J, Danielson E, Buring JE, Manson JE . A randomized factorial trial of vitamins C and E and beta carotene in the secondary prevention of cardiovascular events in women: results from the Women's Antioxidant Cardiovascular Study. Arch Intern Med. 2007; 167: 1610–1618.CrossrefMedlineGoogle Scholar
    • 157. Chen XL, Dodd G, Thomas S, Zhang X, Wasserman MA, Rovin BH, Kunsch C . Activation of Nrf2/ARE pathway protects endothelial cells from oxidant injury and inhibits inflammatory gene expression. Am J Physiol Heart Circ Physiol. 2006; 290: H1862–H1870.CrossrefMedlineGoogle Scholar
    • 158. Levonen AL, Inkala M, Heikura T, Jauhiainen S, Jyrkkanen HK, Kansanen E, Maatta K, Romppanen E, Turunen P, Rutanen J, Yla-Herttuala S . Nrf2 gene transfer induces antioxidant enzymes and suppresses smooth muscle cell growth in vitro and reduces oxidative stress in rabbit aorta in vivo. Arterioscler Thromb Vasc Biol. 2007; 27: 741–747.LinkGoogle Scholar
    • 159. Velmurugan K, Alam J, McCord JM, Pugazhenthi S . Synergistic induction of heme oxygenase-1 by the components of the antioxidant supplement Protandim. Free Radic Biol Med. 2009; 46: 430–440.CrossrefMedlineGoogle Scholar
    • 160. Sussan TE, Jun J, Thimmulappa R, Bedja D, Antero M, Gabrielson KL, Polotsky VY, Biswal S . Disruption of Nrf2, a key inducer of antioxidant defenses, attenuates ApoE-mediated atherosclerosis in mice. PLoS One. 2008; 3: e3791.CrossrefMedlineGoogle Scholar
    • 161. Spyridopoulos I, Haendeler J, Urbich C, Brummendorf TH, Oh H, Schneider MD, Zeiher AM, Dimmeler S . Statins enhance migratory capacity by upregulation of the telomere repeat-binding factor TRF2 in endothelial progenitor cells. Circulation. 2004; 110: 3136–3142.LinkGoogle Scholar
    • 162. Pernice F, Floccari F, Caccamo C, Belghity N, Mantuano S, Pacile ME, Romeo A, Nostro L, Barilla A, Crasci E, Frisina N, Buemi M . Chromosomal damage and atherosclerosis: a protective effect from simvastatin. Eur J Pharmacol. 2006; 532: 223–229.CrossrefMedlineGoogle Scholar
    • 163. Schupp N, Schmid U, Heidland A, Stopper H . Rosuvastatin protects against oxidative stress and DNA damage in vitro via upregulation of glutathione synthesis. Atherosclerosis. 2008; 199: 278–287.CrossrefMedlineGoogle Scholar
    • 164. Oeseburg H, Iusuf D, van der Harst P, van Gilst WH, Henning RH, Roks AJ . Bradykinin protects against oxidative stress-induced endothelial cell senescence. Hypertension. 2009; 53: 417–422.LinkGoogle Scholar
    • 165. Fontana L, Vinciguerra M, Longo VD . Growth factors, nutrient signaling, and cardiovascular aging. Circ Res. 110: 1139–1150.LinkGoogle Scholar
    • 166. Guo ZM, Yang H, Hamilton ML, VanRemmen H, Richardson A . Effects of age and food restriction on oxidative DNA damage and antioxidant enzyme activities in the mouse aorta. Mech Ageing Dev. 2001; 122: 1771–1786.CrossrefMedlineGoogle Scholar
    • 167. 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: 304–312.CrossrefMedlineGoogle Scholar
    • 168. Zanetti M, Gortan Cappellari G, Burekovic I, Barazzoni R, Stebel M, Guarnieri G . Caloric restriction improves endothelial dysfunction during vascular aging: effects on nitric oxide synthase isoforms and oxidative stress in rat aorta. Exp Gerontol. 2010; 45: 848–855.CrossrefMedlineGoogle Scholar
    • 169. Fornieri C, Taparelli F, Quaglino D, Contri MB, Davidson JM, Algeri S, Ronchetti IP . The effect of caloric restriction on the aortic tissue of aging rats. Connect Tissue Res. 1999; 40: 131–143.CrossrefMedlineGoogle Scholar
    • 170. Ungvari Z, Parrado-Fernandez C, Csiszar A, de Cabo R . Mechanisms underlying caloric restriction and lifespan regulation: implications for vascular aging. Circ Res. 2008; 102: 519–528.LinkGoogle Scholar
    • 171. Yang H, Shi M, Story J, Richardson A, Guo Z . Food restriction attenuates age-related increase in the sensitivity of endothelial cells to oxidized lipids. J Gerontol A Biol Sci Med Sci. 2004; 59: 316–323.CrossrefMedlineGoogle Scholar
    • 172. Shinmura K . Cardiovascular protection afforded by caloric restriction: essential role of nitric oxide synthase. Geriatr Gerontol Int. 2011; 11: 143–156.CrossrefMedlineGoogle Scholar
    • 173. Csiszar A, Labinskyy N, Jimenez R, Pinto JT, Ballabh P, Losonczy G, Pearson KJ, de Cabo R, Ungvari Z . Anti-oxidative and anti-inflammatory vasoprotective effects of caloric restriction in aging: role of circulating factors and SIRT1. Mech Ageing Dev. 2009; 130: 518–527.CrossrefMedlineGoogle Scholar
    • 174. Oellerich M, Potente M . FOXOs and sirtuins in vascular growth, maintenance, and aging. Circ Res. 2012; 110: 1238–1251.LinkGoogle Scholar
    • 175. Zhang QJ, Wang Z, Chen HZ, Zhou S, Zheng W, Liu G, Wei YS, Cai H, Liu DP, Liang CC . Endothelium-specific overexpression of class III deacetylase SIRT1 decreases atherosclerosis in apolipoprotein E-deficient mice. Cardiovasc Res. 2008; 80: 191–199.CrossrefMedlineGoogle Scholar
    • 176. Mattagajasingh I, Kim CS, Naqvi A, Yamamori T, Hoffman TA, Jung SB, DeRicco J, Kasuno K, Irani K . SIRT1 promotes endothelium-dependent vascular relaxation by activating endothelial nitric oxide synthase. Proc Natl Acad Sci U S A. 2007; 104: 14855–14860.CrossrefMedlineGoogle Scholar
    • 177. Ota H, Akishita M, Eto M, Iijima K, Kaneki M, Ouchi Y . Sirt1 modulates premature senescence-like phenotype in human endothelial cells. J Mol Cell Cardiol. 2007; 43: 571–579.CrossrefMedlineGoogle Scholar
    • 178. Chen Z, Peng IC, Cui X, Li YS, Chien S, Shyy JY . Shear stress, SIRT1, and vascular homeostasis. Proc Natl Acad Sci U S A. 107: 10268–10273.CrossrefMedlineGoogle Scholar
    • 179. Stein S, Lohmann C, Schafer N, Hofmann J, Rohrer L, Besler C, Rothgiesser KM, Becher B, Hottiger MO, Boren J, McBurney MW, Landmesser U, Luscher TF, Matter CM . SIRT1 decreases Lox-1-mediated foam cell formation in atherogenesis. Eur Heart J. 2010; 31: 2301–2309.CrossrefMedlineGoogle Scholar
    • 180. Pearson KJ, Baur JA, Lewis KN , et al . Resveratrol delays age-related deterioration and mimics transcriptional aspects of dietary restriction without extending life span. Cell Metab. 2008; 8: 157–168.CrossrefMedlineGoogle Scholar
    • 181. Huang JY, Hirschey MD, Shimazu T, Ho L, Verdin E . Mitochondrial sirtuins. Biochim Biophys Acta. 2010; 1804: 1645–1651.CrossrefMedlineGoogle Scholar
    • 182. Verdin E, Hirschey MD, Finley LW, Haigis MC . Sirtuin regulation of mitochondria: energy production, apoptosis, and signaling. Trends Biochem Sci. 2010; 35: 669–675.CrossrefMedlineGoogle Scholar

    eLetters(0)

    eLetters should relate to an article recently published in the journal and are not a forum for providing unpublished data. Comments are reviewed for appropriate use of tone and language. Comments are not peer-reviewed. Acceptable comments are posted to the journal website only. Comments are not published in an issue and are not indexed in PubMed. Comments should be no longer than 500 words and will only be posted online. References are limited to 10. Authors of the article cited in the comment will be invited to reply, as appropriate.

    Comments and feedback on AHA/ASA Scientific Statements and Guidelines should be directed to the AHA/ASA Manuscript Oversight Committee via its Correspondence page.